1
|
Santambrogio L. Haptenization as the missing link between vasculitis and myeloperoxidase. J Clin Invest 2025; 135:e191587. [PMID: 40231470 PMCID: PMC11996909 DOI: 10.1172/jci191587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
A wide variety of medications can induce adverse immune events and autoimmune responses such as vasculitis. Mechanistically, small molecule drugs known as haptens bind and modify endogenous proteins, triggering such immune reactions. In this issue of the JCI, Xi and colleagues investigated the immunological mechanism of autoimmune vasculitis associated with hydralazine. Notably, hydralazine-based haptenization modified myeloperoxidase (MPO), inducing the enzyme conformational change. The hydralazine-modified MPO induced IgM antibody specific for the modified enzyme, followed by immune complex precipitation, tissue deposition, and complement activation. These findings provide a mechanism by which hydralazine induces a type III hypersensitivity reaction associated with mild to severe vasculitis. The study serves as an example for understanding haptenation and may inform the development of diagnostics for determining susceptibility to drug-induced allergic or autoimmune responses.
Collapse
|
2
|
Hu H, Vomund AN, Peterson OJ, Srivastava N, Li T, Kain L, Beatty WL, Zhang B, Hsieh CS, Teyton L, Lichti CF, Unanue ER, Wan X. Crinophagic granules in pancreatic β cells contribute to mouse autoimmune diabetes by diversifying pathogenic epitope repertoire. Nat Commun 2024; 15:8318. [PMID: 39333495 PMCID: PMC11437215 DOI: 10.1038/s41467-024-52619-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 09/13/2024] [Indexed: 09/29/2024] Open
Abstract
Autoimmune attack toward pancreatic β cells causes permanent loss of glucose homeostasis in type 1 diabetes (T1D). Insulin secretory granules store and secrete insulin but are also thought to be tissue messengers for T1D. Here, we show that the crinophagic granules (crinosome), a minor set of vesicles formed by fusing lysosomes with the conventional insulin dense-core granules (DCG), are pathogenic in T1D development in mouse models. Pharmacological inhibition of crinosome formation in β cells delays T1D progression without affecting the dominant DCGs. Mechanistically, crinophagy inhibition diminishes the epitope repertoire in pancreatic islets, including cryptic, modified and disease-relevant epitopes derived from insulin. These unconventional insulin epitopes are largely undetectable in the MHC-II epitope repertoire of the thymus, where only canonical insulin epitopes are presented. CD4+ T cells targeting unconventional insulin epitopes display autoreactive phenotypes, unlike tolerized T cells recognizing epitopes presented in the thymus. Thus, the crinophagic pathway emerges as a tissue-intrinsic mechanism that transforms insulin from a signature thymic self-protein to a critical autoantigen by creating a peripheral-thymic mismatch in the epitope repertoire.
Collapse
Affiliation(s)
- Hao Hu
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anthony N Vomund
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Orion J Peterson
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Neetu Srivastava
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Tiandao Li
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lisa Kain
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Wandy L Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bo Zhang
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Chyi-Song Hsieh
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Luc Teyton
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Cheryl F Lichti
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Emil R Unanue
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoxiao Wan
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3
|
Kelly JJ, Bloodworth N, Shao Q, Shabanowitz J, Hunt D, Meiler J, Pires MM. A Chemical Approach to Assess the Impact of Post-translational Modification on MHC Peptide Binding and Effector Cell Engagement. ACS Chem Biol 2024; 19:1991-2001. [PMID: 39150956 PMCID: PMC11420952 DOI: 10.1021/acschembio.4c00312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/31/2024] [Accepted: 08/08/2024] [Indexed: 08/18/2024]
Abstract
The human major histocompatibility complex (MHC) plays a pivotal role in the presentation of peptidic fragments from proteins, which can originate from self-proteins or from nonhuman antigens, such as those produced by viruses or bacteria. To prevent cytotoxicity against healthy cells, thymocytes expressing T cell receptors (TCRs) that recognize self-peptides are removed from circulation (negative selection), thus leaving T cells that recognize nonself-peptides. Current understanding suggests that post-translationally modified (PTM) proteins and the resulting peptide fragments they generate following proteolysis are largely excluded from negative selection; this feature means that PTMs can generate nonself-peptides that potentially contribute to the development of autoreactive T cells and subsequent autoimmune diseases. Although it is well-established that PTMs are prevalent in peptides present on MHCs, the precise mechanisms by which PTMs influence the antigen presentation machinery remain poorly understood. In the present work, we introduce chemical modifications mimicking PTMs on synthetic peptides. This is the first systematic study isolating the impact of PTMs on MHC binding and also their impact on TCR recognition. Our findings reveal various ways PTMs alter antigen presentation, which could have implications for tumor neoantigen presentation.
Collapse
Affiliation(s)
- Joey J. Kelly
- Department
of Chemistry University of Virginia Charlottesville, Virginia 22904, United States
| | - Nathaniel Bloodworth
- Division
of Clinical Pharmacology, Department of MedicineVanderbilt University Medical Center, Nashville, Tennessee 37240, United States
| | - Qianqian Shao
- Department
of Chemistry University of Virginia Charlottesville, Virginia 22904, United States
| | - Jeffrey Shabanowitz
- Department
of Chemistry University of Virginia Charlottesville, Virginia 22904, United States
| | - Donald Hunt
- Department
of Chemistry University of Virginia Charlottesville, Virginia 22904, United States
| | - Jens Meiler
- Division
of Clinical Pharmacology, Department of MedicineVanderbilt University Medical Center, Nashville, Tennessee 37240, United States
- Institute
of Drug Discovery, Faculty of MedicineUniversity
of Leipzig, Leipzig, SAC 04103, Germany
- Center
for Structural Biology Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Chemistry Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Marcos M. Pires
- Department
of Chemistry University of Virginia Charlottesville, Virginia 22904, United States
| |
Collapse
|
4
|
Nanaware PP, Khan ZN, Clement CC, Shetty M, Mota I, Seltzer ES, Dzieciatkowska M, Gamboni F, D'Alessandro A, Ng C, Nagayama M, Lichti CF, Soni RK, Jacob B Geri, Matei I, Lyden D, Longman R, Lu TT, Wan X, Unanue ER, Stern LJ, Santambrogio L. Role of the afferent lymph as an immunological conduit to analyze tissue antigenic and inflammatory load. Cell Rep 2024; 43:114311. [PMID: 38848214 PMCID: PMC11233987 DOI: 10.1016/j.celrep.2024.114311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 06/09/2024] Open
Abstract
The lymphatic fluid is the conduit by which part of the tissue "omics" is transported to the draining lymph node for immunosurveillance. Following cannulation of the pre-nodal cervical and mesenteric afferent lymphatics, herein we investigate the lymph proteomic composition, uncovering that its composition varies according to the tissue of origin. Tissue specificity is also reflected in the dendritic cell-major histocompatibility complex class II-eluted immunopeptidome harvested from the cervical and mesenteric nodes. Following inflammatory disruption of the gut barrier, the lymph antigenic and inflammatory loads are analyzed in both mice and subjects with inflammatory bowel diseases. Gastrointestinal tissue damage reflects the lymph inflammatory and damage-associated molecular pattern signatures, microbiome-derived by-products, and immunomodulatory molecules, including metabolites of the gut-brain axis, mapped in the afferent mesenteric lymph. Our data point to the relevance of the lymphatic fluid to probe the tissue-specific antigenic and inflammatory load transported to the draining lymph node for immunosurveillance.
Collapse
Affiliation(s)
- Padma P Nanaware
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Zohaib N Khan
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Cristina C Clement
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Madhur Shetty
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ines Mota
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ethan S Seltzer
- Pediatric Rheumatology and Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York NY 100021, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Charles Ng
- Department of Pathology and Laboratory Medicine, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY 10065, USA
| | - Manabu Nagayama
- Division of Gastroenterology and Hepatology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY 10065, USA
| | - Cheryl F Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Rajesh K Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York 10032, NY, USA
| | - Jacob B Geri
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| | - David Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| | - Randy Longman
- Division of Gastroenterology and Hepatology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY 10065, USA
| | - Theresa T Lu
- Pediatric Rheumatology and Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York NY 100021, USA
| | - Xiaoxiao Wan
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Lawrence J Stern
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY 10065, USA.
| |
Collapse
|
5
|
Zacarias O, Clement CC, Cheng SY, Rosas M, Gonzalez C, Peter M, Coopman P, Champeil E. Mitomycin C and its analog trigger cytotoxicity in MCF-7 and K562 cancer cells through the regulation of RAS and MAPK/ERK pathways. Chem Biol Interact 2024; 395:111007. [PMID: 38642817 PMCID: PMC11102841 DOI: 10.1016/j.cbi.2024.111007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/27/2024] [Accepted: 04/14/2024] [Indexed: 04/22/2024]
Abstract
Mitomycin C (MC) is an anti-cancer drug which functions by forming interstrand crosslinks (ICLs) between opposing DNA strands. MC analog, 10-decarbamoyl mitomycin C (DMC), unlike MC, has stronger cytotoxic effects on cancer cells with TP53 mutation. We previously demonstrated that MC/DMC could activate p21WAF1/CIP1 in MCF-7 (TP53-proficient) and K562 (TP53 deficient) cells in a TP53-independent mode. We also found that MC/DMC regulate AKT activation in a TP53-dependent manner and that AKT deactivation is not associated with the activation of p21WAF1/CIP1 in response to MC/DMC treatment. RAS proteins are known players in the upstream mediated signaling of p21WAF1/CIP1 activation that leads to control of cell proliferation and cell death. Thus, this prompted us to investigate the effect of both drugs on the expression of RAS proteins and regulation of the MAPK/ERK signaling pathways in MCF-7 and K562 cancer cells. To accomplish this goal, we performed comparative label free proteomics profiling coupled to bioinformatics/complementary phosphoprotein arrays and Western blot validations of key signaling molecules. The MAPK/ERK pathway exhibited an overall downregulation upon MC/DMC treatment in MCF-7 cells but only DMC exhibited a mild downregulation of that same pathway in TP53 mutant K562 cells. Furthermore, treatment of MCF-7 and K562 cell lines with oligonucleotides containing the interstrand crosslinks (ICLs) formed by MC or DMC shows that both ICLs had a stronger effect on the downregulation of RAS protein expression in mutant TP53 K562 cells. We discuss the implication of this regulation of the MAPK/ERK pathway in relation to cellular TP53 status.
Collapse
Affiliation(s)
- Owen Zacarias
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA
| | - Cristina C Clement
- Radiation Oncology Department, Weill Cornell Medicine, New York, New York, 10065, USA.
| | - Shu-Yuan Cheng
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA; Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, 10016, USA.
| | - Melissa Rosas
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA
| | - Christina Gonzalez
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA
| | - Marion Peter
- IRCM, University Montpellier, ICM, INSERM, CNRS, Campus Val d'Aurelle, 208 avenue des apothicaires, 34298, Montpellier, Cédex 5, France
| | - Peter Coopman
- IRCM, University Montpellier, ICM, INSERM, CNRS, Campus Val d'Aurelle, 208 avenue des apothicaires, 34298, Montpellier, Cédex 5, France
| | - Elise Champeil
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA; Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, USA.
| |
Collapse
|
6
|
Stern LJ, Clement C, Galluzzi L, Santambrogio L. Non-mutational neoantigens in disease. Nat Immunol 2024; 25:29-40. [PMID: 38168954 PMCID: PMC11075006 DOI: 10.1038/s41590-023-01664-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/29/2023] [Indexed: 01/05/2024]
Abstract
The ability of mammals to mount adaptive immune responses culminating with the establishment of immunological memory is predicated on the ability of the mature T cell repertoire to recognize antigenic peptides presented by syngeneic MHC class I and II molecules. Although it is widely believed that mature T cells are highly skewed towards the recognition of antigenic peptides originating from genetically diverse (for example, foreign or mutated) protein-coding regions, preclinical and clinical data rather demonstrate that novel antigenic determinants efficiently recognized by mature T cells can emerge from a variety of non-mutational mechanisms. In this Review, we describe various mechanisms that underlie the formation of bona fide non-mutational neoantigens, such as epitope mimicry, upregulation of cryptic epitopes, usage of non-canonical initiation codons, alternative RNA splicing, and defective ribosomal RNA processing, as well as both enzymatic and non-enzymatic post-translational protein modifications. Moreover, we discuss the implications of the immune recognition of non-mutational neoantigens for human disease.
Collapse
Affiliation(s)
- Lawrence J Stern
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbiology Program, UMass Chan Medical School, Worcester, MA, USA
| | - Cristina Clement
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Chiriţoiu GN, Munteanu CV, Şulea TA, Spiridon L, Petrescu AJ, Jandus C, Romero P, Petrescu ŞM. Methionine oxidation selectively enhances T cell reactivity against a melanoma antigen. iScience 2023; 26:107205. [PMID: 37485346 PMCID: PMC10362274 DOI: 10.1016/j.isci.2023.107205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/02/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023] Open
Abstract
The impact of the peptide amino acids side-chain modifications on the immunological recognition has been scarcely explored. We investigate here the effect of methionine oxidation on the antigenicity of the melanoma immunodominant peptide 369-YMDGTMSQV-377 (YMD). Using CD8+ T cell activation assays, we found that the antigenicity of the sulfoxide form is higher when compared to the YMD peptide. This is consistent with free energy computations performed on HLA-A∗02:01/YMD/TCR complex showing that this is lowered upon oxidation, paired with a steep increase in order at atomic level. Oxidized YMD forms were identified at the melanoma cell surface by LC-MS/MS analysis. These results demonstrate that methionine oxidation in the antigenic peptides may generate altered peptide ligands with increased antigenicity, and that this oxidation may occur in vivo, opening up the possibility that high-affinity CD8+ T cells might be naturally primed in the course of melanoma progression, as a result of immunosurveillance.
Collapse
Affiliation(s)
- Gabriela N. Chiriţoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Splaiul Independenței 296, 060031 Bucharest, Romania
| | - Cristian V.A. Munteanu
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry, Splaiul Independenței 296, 060031 Bucharest, Romania
| | - Teodor A. Şulea
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry, Splaiul Independenței 296, 060031 Bucharest, Romania
| | - Laurenţiu Spiridon
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry, Splaiul Independenței 296, 060031 Bucharest, Romania
| | - Andrei-Jose Petrescu
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry, Splaiul Independenței 296, 060031 Bucharest, Romania
| | - Camilla Jandus
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Pedro Romero
- Departement of Oncology, UNIL-CHUV, University of Lausanne, Epalinges, Switzerland
| | - Ştefana M. Petrescu
- Department of Molecular Cell Biology, Institute of Biochemistry, Splaiul Independenței 296, 060031 Bucharest, Romania
| |
Collapse
|
8
|
Santambrogio L. Autoimmunity to the modified self. Science 2023; 379:1092-1093. [PMID: 36927028 DOI: 10.1126/science.adg3925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Protein posttranslational modifications can break tolerance to the self-proteome.
Collapse
Affiliation(s)
- Laura Santambrogio
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
9
|
Santambrogio L, Franco A. The yin/yang balance of the MHC-self -immunopeptidome. Front Immunol 2022; 13:1035363. [PMID: 36405763 PMCID: PMC9666884 DOI: 10.3389/fimmu.2022.1035363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/07/2022] [Indexed: 07/22/2023] Open
Abstract
The MHC-self immunopeptidome of professional antigen presenting cells is a cognate ligand for the TCRs expressed on both conventional and thymic-derived natural regulatory T cells. In regulatory T cells, the TCR signaling associated with MHC-peptide recognition induces antigen specific as well as bystander immunosuppression. On the other hand, TCR activation of conventional T cells is associated with protective immunity. As such the peripheral T cell repertoire is populated by a number of T cells with different phenotypes and different TCRs, which can recognize the same MHC-self-peptide complex, resulting in opposite immunological outcomes. This article summarizes what is known about regulatory and conventional T cell recognition of the MHC-self-immunopeptidome at steady state and in inflammatory conditions associated with increased T and B cell self-reactivity, discussing how changes in the MHC-ligandome including epitope copy number and post-translational modifications can tilt the balance toward the expansion of pro-inflammatory or regulatory T cells.
Collapse
Affiliation(s)
- Laura Santambrogio
- Department of Radiation Oncology, Physiology and Biophysics, Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Alessandra Franco
- University of California San Diego School of Medicine, Department of Pediatrics, La Jolla, CA, United States
| |
Collapse
|
10
|
Ma X, Sun Y, Wang D, Lin Y, Chang H. Blood transthyretin for predicting immunoglobulin A vasculitis nephritis outcome in children. Int Immunopharmacol 2022; 108:108765. [PMID: 35397389 DOI: 10.1016/j.intimp.2022.108765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND IgA vasculitis is characterized by inflammation of the blood vessels, which can result in microvascular destruction and consequently renal damage. Transthyretin is a newly discovered angiogenesis regulator in promoting microvascular regeneration. This indicates that transthyretin may act as a potential predictor of IgAV as well as IgAVN. METHODS This retrospective study included 125 patients newly diagnosed as IgAV with demographic and laboratory parameters. Of these, 78 patients had demonstrated internal organ damage and 47 patients with only skin and joint injury. Of 78 patients with organ impairment, 27 were diagnosed of renal involvement. Then we evaluated the relationship between NLR, total protein, albumin, globulin, transthyretin, B lymphocyte counts and the severity of IgAV. RESULTS For patients with internal organ or renal involvement, the level of transthyretin were lower than non-internal organ damage group (p < 0.001 for both group). Remarkably, the NLR was only higher in patients with internal organ damage group (p = 0.019). Logistic regression analysis showed that NLR and transthyretin both were risk factors for internal organ involvement (OR = 1.768, 0.973 separately), and only transthyretin is the independent risk for renal involvement (OR = 0.981, p < 0.05). The ROC analysis showed an AUC of 0.626 for NLR, 0.815 for transthyretin in predicting organ damage, 0.755 for transthyretin in patients with renal involvement (p < 0.05, to all parameters). CONCLUSIONS Transthyretin is a better predictor in predicting internal organ or renal involvement than NLR, and low plasma transthyretin concentration can increase the risk of renal involvement in IgAV patients.
Collapse
Affiliation(s)
- Xiancheng Ma
- Pediatrics Department, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Yunxiao Sun
- Pediatrics Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, China
| | - Dahai Wang
- Pediatrics Department, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Yi Lin
- Pediatrics Department, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Hong Chang
- Pediatrics Department, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China.
| |
Collapse
|
11
|
Wieczorek E, Ożyhar A. Transthyretin: From Structural Stability to Osteoarticular and Cardiovascular Diseases. Cells 2021; 10:1768. [PMID: 34359938 PMCID: PMC8307983 DOI: 10.3390/cells10071768] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/29/2021] [Accepted: 07/09/2021] [Indexed: 01/10/2023] Open
Abstract
Transthyretin (TTR) is a tetrameric protein transporting hormones in the plasma and brain, which has many other activities that have not been fully acknowledged. TTR is a positive indicator of nutrition status and is negatively correlated with inflammation. TTR is a neuroprotective and oxidative-stress-suppressing factor. The TTR structure is destabilized by mutations, oxidative modifications, aging, proteolysis, and metal cations, including Ca2+. Destabilized TTR molecules form amyloid deposits, resulting in senile and familial amyloidopathies. This review links structural stability of TTR with the environmental factors, particularly oxidative stress and Ca2+, and the processes involved in the pathogenesis of TTR-related diseases. The roles of TTR in biomineralization, calcification, and osteoarticular and cardiovascular diseases are broadly discussed. The association of TTR-related diseases and vascular and ligament tissue calcification with TTR levels and TTR structure is presented. It is indicated that unaggregated TTR and TTR amyloid are bound by vicious cycles, and that TTR may have an as yet undetermined role(s) at the crossroads of calcification, blood coagulation, and immune response.
Collapse
Affiliation(s)
- Elżbieta Wieczorek
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland;
| | | |
Collapse
|
12
|
Pleiotropic consequences of metabolic stress for the major histocompatibility complex class II molecule antigen processing and presentation machinery. Immunity 2021; 54:721-736.e10. [PMID: 33725478 DOI: 10.1016/j.immuni.2021.02.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/30/2020] [Accepted: 02/24/2021] [Indexed: 01/11/2023]
Abstract
Hyperglycemia and hyperlipidemia are often observed in individuals with type II diabetes (T2D) and related mouse models. One dysmetabolic biochemical consequence is the non-enzymatic reaction between sugars, lipids, and proteins, favoring protein glycation, glycoxidation, and lipoxidation. Here, we identified oxidative alterations in key components of the major histocompatibility complex (MHC) class II molecule antigen processing and presentation machinery in vivo under conditions of hyperglycemia-induced metabolic stress. These modifications were linked to epitope-specific changes in endosomal processing efficiency, MHC class II-peptide binding, and DM editing activity. Moreover, we observed some quantitative and qualitative changes in the MHC class II immunopeptidome of Ob/Ob mice on a high-fat diet compared with controls, including changes in the presentation of an apolipoprotein B100 peptide associated previously with T2D and metabolic syndrome-related clinical complications. These findings highlight a link between glycation reactions and altered MHC class II antigen presentation that may contribute to T2D complications.
Collapse
|
13
|
Marx A, Yamada Y, Simon-Keller K, Schalke B, Willcox N, Ströbel P, Weis CA. Thymus and autoimmunity. Semin Immunopathol 2021; 43:45-64. [PMID: 33537838 PMCID: PMC7925479 DOI: 10.1007/s00281-021-00842-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
The thymus prevents autoimmune diseases through mechanisms that operate in the cortex and medulla, comprising positive and negative selection and the generation of regulatory T-cells (Tregs). Egress from the thymus through the perivascular space (PVS) to the blood is another possible checkpoint, as shown by some autoimmune/immunodeficiency syndromes. In polygenic autoimmune diseases, subtle thymic dysfunctions may compound genetic, hormonal and environmental cues. Here, we cover (a) tolerance-inducing cell types, whether thymic epithelial or tuft cells, or dendritic, B- or thymic myoid cells; (b) tolerance-inducing mechanisms and their failure in relation to thymic anatomic compartments, and with special emphasis on human monogenic and polygenic autoimmune diseases and the related thymic pathologies, if known; (c) polymorphisms and mutations of tolerance-related genes with an impact on positive selection (e.g. the gene encoding the thymoproteasome-specific subunit, PSMB11), promiscuous gene expression (e.g. AIRE, PRKDC, FEZF2, CHD4), Treg development (e.g. SATB1, FOXP3), T-cell migration (e.g. TAGAP) and egress from the thymus (e.g. MTS1, CORO1A); (d) myasthenia gravis as the prototypic outcome of an inflamed or disordered neoplastic ‘sick thymus’.
Collapse
Affiliation(s)
- Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Yosuke Yamada
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, 606-8507, Japan
| | - Katja Simon-Keller
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Berthold Schalke
- Department of Neurology, Bezirkskrankenhaus, University of Regensburg, 93042, Regensburg, Germany
| | - Nick Willcox
- Neurosciences Group, Nuffield Department of Clinical Neurology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, University of Göttigen, 37075, Göttingen, Germany
| | - Cleo-Aron Weis
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
14
|
Abstract
The diseases affecting the retina or uvea (iris, ciliary body, or choroid) generate changes in the biochemical or protein composition of ocular fluids/tissues due to disruption of blood-retinal barrier. Ocular infections and inflammations are sight-threatening diseases associated with various infectious and non-infectious etiologies. Several etiological entities cause uveitis, a complex intraocular inflammatory disease. These causes of uveitis differ in different populations due to geographical, racial, and socioeconomic variations. While clinical appearance is sufficiently diagnostic in many diseases, some of the uveitic entities manifest nonspecific or atypical clinical presentation. Identification of biomarkers in such diseases is an important aid in their diagnostic armamentarium. Different diseases and their different severity states release varying concentrations of proteins, which can serve as biomarkers. Proteomics is a high throughput technology and a powerful screening tool for serum biomarkers in various diseases that identifies proteins by mass spectrometry and helps to improve the understanding of pathogenesis of a disease. Proteins determine the biological state of a cell. Once identified as biomarkers, they serve as future diagnostic and pharmaceutical targets. With a potential to redirect the diagnosis of idiopathic uveitis, ocular proteomics provide a new insight into the pathophysiology and therapeutics of various ocular inflammatory diseases. Tears, aqueous and vitreous humor represent potential repositories for proteomic biomarkers discovery in uveitis. With an extensive proteomics work done on animal models of uveitis, various types of human uveitis are being subjected to proteome analysis for biomarker discovery in different ocular fluids (vitreous, aqueous, or tears).
Collapse
Affiliation(s)
- Reema Bansal
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Amod Gupta
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
15
|
Aghamollaei H, Parvin S, Shahriary A. Review of proteomics approach to eye diseases affecting the anterior segment. J Proteomics 2020; 225:103881. [PMID: 32565161 DOI: 10.1016/j.jprot.2020.103881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 01/12/2023]
Abstract
Visual impairment and blindness is a major health burden worldwide, and major ocular diseases causing visual impairment pertain to the anterior segment of the eye. Anterior segment ocular diseases are common, yet complex entities. Although many treatment options and surgical techniques are available for these ailments, the underlying cause and pathogenesis is still unclear. Finding ways to fundamentally treat these patients and rectify the underlying dysregulations leading to the disease may help cure patients completely without major complications. Proteomics approaches are a novel way to distinguish dysregulated proteins in a variety of biological tissues in a hypothesis-free manner, thus helping to find the responsible pathways leading to a certain disease. The aim of the current study is to review the available knowledge in scientific literature regarding the proteomics studies done on anterior segment eye diseases and suggest potential clinical implications to exploit the results of these studies. SIGNIFICANCE: Anterior segment ocular diseases are responsible for a major proportion of visual impairment and blindness worldwide. Although ophthalmologists have several treatment options that can alleviate or control the progression of these diseases, no definite cure is available for most of them. Moreover, because these diseases are progressive, prompt diagnosis is of utmost important. Proteomics studies enable us to identify and quantify the dysregulated proteins in a biological specimen in a hypothesis-free manner. Understanding the dysregulated protein pathways shines a light on the pathogenesis of the disease. Moreover, these dysregulated proteins may act as biomarkers to help in diagnosis and treatment follow-up. Hence, in this article we sought out to review the available scientific literature regarding the proteomics studies of anterior segment ocular diseases and to identify potential applications of proteomic studies in clinic.
Collapse
Affiliation(s)
- Hossein Aghamollaei
- Chemical Injuries Research Center, Systems biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shahram Parvin
- Chemical Injuries Research Center, Systems biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Tomofuji Y, Takaba H, Suzuki HI, Benlaribi R, Martinez CDP, Abe Y, Morishita Y, Okamura T, Taguchi A, Kodama T, Takayanagi H. Chd4 choreographs self-antigen expression for central immune tolerance. Nat Immunol 2020; 21:892-901. [PMID: 32601470 DOI: 10.1038/s41590-020-0717-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Autoreactive T cells are eliminated in the thymus to prevent autoimmunity by promiscuous expression of tissue-restricted self-antigens in medullary thymic epithelial cells. This expression is dependent on the transcription factor Fezf2, as well as the transcriptional regulator Aire, but the entire picture of the transcriptional program has been obscure. Here, we found that the chromatin remodeler Chd4, also called Mi-2β, plays a key role in the self-antigen expression in medullary thymic epithelial cells. To maximize the diversity of self-antigen expression, Fezf2 and Aire utilized completely distinct transcriptional mechanisms, both of which were under the control of Chd4. Chd4 organized the promoter regions of Fezf2-dependent genes, while contributing to the Aire-mediated induction of self-antigens via super-enhancers. Mice deficient in Chd4 specifically in thymic epithelial cells exhibited autoimmune phenotypes, including T cell infiltration. Thus, Chd4 plays a critical role in integrating Fezf2- and Aire-mediated gene induction to establish central immune tolerance.
Collapse
Affiliation(s)
- Yoshihiko Tomofuji
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Takaba
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi I Suzuki
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Molecular Oncology, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rayene Benlaribi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Cristian David Peña Martinez
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Abe
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Morishita
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Section of Animal Models, National Center for Global Health and Medicine, Tokyo, Japan
| | - Akashi Taguchi
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Kodama
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
17
|
Unanue ER, Wan X. The Immunoreactive Platform of the Pancreatic Islets Influences the Development of Autoreactivity. Diabetes 2019; 68:1544-1551. [PMID: 31331989 PMCID: PMC6692819 DOI: 10.2337/dbi18-0048] [Citation(s) in RCA: 12] [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: 04/05/2019] [Accepted: 05/27/2019] [Indexed: 01/23/2023]
Abstract
Tissue homeostasis is maintained through a finely tuned balance between the immune system and the organ-resident cells. Disruption of this process not only results in organ dysfunction but also may trigger detrimental autoimmune responses. The islet of Langerhans consists of the insulin-producing β-cells essential for proper control of body metabolism, but less appreciated is that these cells naturally interact with the immune system, forming a platform by which the β-cell products are sensed, processed, and responded to by the local immune cells, particularly the islet-resident macrophages. Although its physiological outcomes are not completely understood, this immunoreactive platform is crucial for precipitating islet autoreactivity in individuals carrying genetic risks, leading to the development of type 1 diabetes. In this Perspective, we summarize recent studies that examine the cross talk between the β-cells and various immune components, with a primary focus on discussing how antigenic information generated during normal β-cell catabolism can be delivered to the resident macrophage and further recognized by the adaptive CD4 T-cell system, a critical step to initiate autoimmune diabetes. The core nature of the islet immune platform can be extrapolated to other endocrine tissues and may represent a common mechanism underlying the development of autoimmune syndromes influencing multiple endocrine organs.
Collapse
Affiliation(s)
- Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Xiaoxiao Wan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
18
|
Araman C, 't Hart BA. Neurodegeneration meets immunology - A chemical biology perspective. Bioorg Med Chem 2019; 27:1911-1924. [PMID: 30910473 DOI: 10.1016/j.bmc.2019.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 11/16/2022]
Affiliation(s)
- C Araman
- Leiden Institute of Chemistry and the Institute for Chemical Immunology, Leiden University, Leiden, The Netherlands.
| | - B A 't Hart
- University of Groningen, Department of Biomedical Sciences of Cells and Systems, University Medical Centre, Groningen, The Netherlands; Department Anatomy and Neuroscience, Free University Medical Center (VUmc), Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Mahmud SA, Binstadt BA. Autoantibodies in the Pathogenesis, Diagnosis, and Prognosis of Juvenile Idiopathic Arthritis. Front Immunol 2019; 9:3168. [PMID: 30693002 PMCID: PMC6339949 DOI: 10.3389/fimmu.2018.03168] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/24/2018] [Indexed: 12/26/2022] Open
Abstract
Autoantibody production occurs in juvenile idiopathic arthritis (JIA) and numerous other autoimmune diseases. In some conditions, the autoantibodies are clearly pathogenic, whereas in others the roles are less defined. Here we review various autoantibodies associated with JIA, with a particular focus on antinuclear antibodies and antibodies recognizing citrullinated self-antigens. We explore potential mechanisms that lead to the development of autoantibodies and the use of autoantibody testing in diagnosis and prognosis. Finally, we compare and contrast JIA-associated autoantibodies with those found in adults with rheumatoid arthritis (RA).
Collapse
Affiliation(s)
- Shawn A Mahmud
- Division of Pediatric Rheumatology, Department of Pediatrics, and the Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Bryce A Binstadt
- Division of Pediatric Rheumatology, Department of Pediatrics, and the Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
20
|
Cheng SY, Vargas A, Lee JY, Clement CC, Champeil E. Involvement of Akt in mitomycin C and its analog triggered cytotoxicity in MCF-7 and K562 cancer cells. Chem Biol Drug Des 2018; 92:2022-2034. [PMID: 30091208 PMCID: PMC6251731 DOI: 10.1111/cbdd.13374] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/13/2018] [Accepted: 07/22/2018] [Indexed: 01/13/2023]
Abstract
Mitomycin C (MC) is a well-known DNA alkylating agent. MC analog, 10-decarbamoyl mitomycin C (DMC), unlike MC, has stronger effects on cancer with p53 mutation. We previously demonstrated that MC/DMC could activate p21WAF1/CIP1 in MCF-7 (p53-proficient) and K562 (p53-deficient) cells in a p53-independent mode. This study aimed to elucidate the upstream signaling pathway of p21WAF1/CIP1 activation triggered by MC/DMC. Besides p53, Akt plays an important role on deactivating p21WAF1/CIP1 . The results showed that MC/DMC inhibited Akt in MCF-7 cells, but not in K562 cells. By knocking down p53, the Akt inhibition in MCF-7 cells was alleviated. This implied that the deactivated Akt caused by MC/DMC was p53-dependent. With Akt activator (SC79), p21WAF1/CIP1 activation triggered by MC/DMC in MCF-7 cells was not reduced. This indicated that Akt inhibition triggered by MC/DMC was not associated with MC/DMC-induced p21WAF1/CIP1 activation. Label-free quantitative proteomic profiling analysis revealed that DMC has a stronger effect on down-regulating the PI3K/Akt signaling pathway in MCF-7 cells as compared to MC. No significant effect of MC/DMC on PI3K/Akt in K562 cells was observed. In summary, MC/DMC regulate Akt activation in a p53-dependent manner. This Akt deactivation is not associated with p21WAF1/CIP1 activation in response to MC/DMC.
Collapse
Affiliation(s)
- Shu-Yuan Cheng
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York City, New York
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York City, New York
| | - Anayatzinc Vargas
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York City, New York
| | - Ji-Young Lee
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York City, New York
| | - Cristina C Clement
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
- Chemistry Department, Lehman College, City University of New York, Bronx, New York
| | - Elise Champeil
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York City, New York
| |
Collapse
|
21
|
Wan X, Unanue ER. Antigen recognition in autoimmune diabetes: a novel pathway underlying disease initiation. PRECISION CLINICAL MEDICINE 2018; 1:102-110. [PMID: 30687564 PMCID: PMC6333048 DOI: 10.1093/pcmedi/pby015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/13/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Development of human autoimmune disorders results from complex interplay among genetic, environmental, and immunological risk factors. Despite much heterogeneity in environmental triggers, the leading genes that give the propensity for tissue-specific autoimmune diseases, such as type 1 diabetes, are those associated with particular class II major histocompatibility complex alleles. Such genetic predisposition precipitates presentation of tissue antigens to MHC-II-restricted CD4 T cells. When properly activated, these self-reactive CD4 T cells migrate to the target tissue and trigger the initial immune attack. Using the non-obese diabetic mouse model of spontaneous autoimmune diabetes, much insight has been gained in understanding how presentation of physiological levels of self-antigens translates into pathological outcomes. In this review, we summarize recent advances illustrating the features of the antigen presenting cells, the sites of the antigen recognition, and the nature of the consequent T cell responses. We emphasize emerging evidence that highlights the importance of systemic presentation of catabolized tissue antigens in mobilization of pathogenic T cells. The implication of these studies in therapeutic perspectives is also discussed.
Collapse
Affiliation(s)
- Xiaoxiao Wan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
22
|
Wong A, Bryzek D, Dobosz E, Scavenius C, Svoboda P, Rapala-Kozik M, Lesner A, Frydrych I, Enghild J, Mydel P, Pohl J, Thompson PR, Potempa J, Koziel J. A Novel Biological Role for Peptidyl-Arginine Deiminases: Citrullination of Cathelicidin LL-37 Controls the Immunostimulatory Potential of Cell-Free DNA. THE JOURNAL OF IMMUNOLOGY 2018; 200:2327-2340. [PMID: 29475987 DOI: 10.4049/jimmunol.1701391] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/24/2018] [Indexed: 01/08/2023]
Abstract
LL-37, the only human cathelicidin that is released during inflammation, is a potent regulator of immune responses by facilitating delivery of oligonucleotides to intracellular TLR-9, thereby enhancing the response of human plasmacytoid dendritic cells (pDCs) to extracellular DNA. Although important for pathogen recognition, this mechanism may facilitate development of autoimmune diseases. In this article, we show that citrullination of LL-37 by peptidyl-arginine deiminases (PADs) hindered peptide-dependent DNA uptake and sensing by pDCs. In contrast, carbamylation of the peptide (homocitrullination of Lys residues) had no effect. The efficiency of LL-37 binding to oligonucleotides and activation of pDCs was found to be inversely proportional to the number of citrullinated residues in the peptide. Similarly, preincubation of carbamylated LL-37 with PAD2 abrogated the peptide's ability to bind DNA. Conversely, LL-37 with Arg residues substituted by homoarginine, which cannot be deiminated, elicited full activity of native LL-37 regardless of PAD2 treatment. Taken together, the data showed that citrullination abolished LL-37 ability to bind DNA and altered the immunomodulatory function of the peptide. Both activities were dependent on the proper distribution of guanidinium side chains in the native peptide sequence. Moreover, our data suggest that cathelicidin/LL-37 is citrullinated by PADs during NET formation, thus affecting the inflammatory potential of NETs. Together this may represent a novel mechanism for preventing the breakdown of immunotolerance, which is dependent on the response of APCs to self-molecules (including cell-free DNA); overactivation may facilitate development of autoimmunity.
Collapse
Affiliation(s)
- Alicia Wong
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387 Krakow, Poland
| | - Danuta Bryzek
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387 Krakow, Poland
| | - Ewelina Dobosz
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387 Krakow, Poland
| | - Carsten Scavenius
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Pavel Svoboda
- Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30329
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387 Krakow, Poland
| | - Adam Lesner
- Faculty of Chemistry, University of Gdansk, 80-309 Gdansk, Poland
| | - Ivo Frydrych
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77126 Olomouc, Czech Republic
| | - Jan Enghild
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Piotr Mydel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387 Krakow, Poland.,Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Jan Pohl
- Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30329
| | - Paul R Thompson
- Department of Biochemistry and Molecular Pharmacology, UMass Medical School, Worcester, MA 01605; and
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387 Krakow, Poland; .,Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, University of Louisville, Louisville, KY 40202
| | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387 Krakow, Poland;
| |
Collapse
|
23
|
Loshaj-Shala A, Colzani M, Brezovska K, Poceva Panovska A, Suturkova L, Beretta G. Immunoproteomic identification of antigenic candidate Campylobacter jejuni and human peripheral nerve proteins involved in Guillain-Barré syndrome. J Neuroimmunol 2018; 317:77-83. [PMID: 29338928 DOI: 10.1016/j.jneuroim.2018.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/04/2018] [Accepted: 01/07/2018] [Indexed: 12/11/2022]
Abstract
Immunoproteomics is become a potent methodology used for identifying immunoreactive proteins. In this study, an immunoproteomic approach based on 2-dimensional gel electrophoresis (2D-PAGE) and immunoblotting combined with high resolution mass spectrometry (MS) was used to identify immunoreactive proteins that might be involved in mechanisms of Guillain-Barré syndrome (GBS) development, regardless of their potential reciprocal molecular mimicry. Proteins isolated from C. jejuni and human peripheral nerve tissue (HPN) were separated with 2D SDS-PAGE and subjected to western blotting using serum samples from GBS patients. The peptides generated after proteolysis of the immunoreactive proteins were submitted to nanoflow-high performance liquid chromatography-nano electrospray ionization coupled to high resolution mass spectrometry (nHPLC-nESI-MS and MS/MS) followed by SEQUESTdata analysis for proteins identification. In C. jejuni, immunoreactivity was found for GroEL and DnaK, structural proteins (MOMP), key enzymatic proteins necessary for the microbial proliferation (adenylate kinase, enolase, inorganic pyrophosphatase and aspartate ammonia-lyase), and antioxidant enzymes (alkyl hydroperoxide reductase-AhpC and DNA protection during starvation protein - DNA protection factor against Fe2+-mediated oxidative stress). HPN immunoreactive proteins identified were heat shock proteins (HSP), intermediate filaments (vimentin and desmin), and other proteins and enzymes such as troponin/tropomyosin complex and ATP synthase subunit beta and the keratan sulfate proteoglycan lumican. The targeting of vimentin and desmin, suggested that the neuronal autoimmune damage is specifically directed to intermediate neuronal (vimentin) and neuromuscular IF, probably localized nearby cell surface, affording increased accessibility to autoantibodies. These findings suggest that the post-infectious development of GBS may be also associated to additional concomitant immune factors that lead to nerve damage generated by auto-immune trigger(s) different from molecular mimicry.
Collapse
Affiliation(s)
- Aida Loshaj-Shala
- Department of Pharmacy, Faculty of Medicine, University Hasan Prishtina, Pristina, Kosovo
| | - Mara Colzani
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Katerina Brezovska
- Faculty of Pharmacy, University Ss. Cyril and Methodius, Skopje, Macedonia
| | | | - Ljubica Suturkova
- Faculty of Pharmacy, University Ss. Cyril and Methodius, Skopje, Macedonia
| | - Giangiacomo Beretta
- Department of Environmental Science and Policy, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
24
|
Matsuzaki T, Akasaki Y, Olmer M, Alvarez‐Garcia O, Reixach N, Buxbaum JN, Lotz MK. Transthyretin deposition promotes progression of osteoarthritis. Aging Cell 2017; 16:1313-1322. [PMID: 28941045 PMCID: PMC5676063 DOI: 10.1111/acel.12665] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2017] [Indexed: 01/01/2023] Open
Abstract
Deposition of amyloid is a common aging-associated phenomenon in several aging-related diseases. Osteoarthritis (OA) is the most prevalent joint disease, and aging is its major risk factor. Transthyretin (TTR) is an amyloidogenic protein that is deposited in aging and OA-affected human cartilage and promotes inflammatory and catabolic responses in cultured chondrocytes. Here, we investigated the role of TTR in vivo using transgenic mice overexpressing wild-type human TTR (hTTR-TG). Although TTR protein was detected in cartilage in hTTR-TG mice, the TTR transgene was highly overexpressed in liver, but not in chondrocytes. OA was surgically induced by destabilizing the medial meniscus (DMM) in hTTR-TG mice, wild-type mice of the same strain (WT), and mice lacking endogenous Ttr genes. In the DMM model, both cartilage and synovitis histological scores were significantly increased in hTTR-TG mice. Further, spontaneous degradation and OA-like changes in cartilage and synovium developed in 18-month-old hTTR mice. Expression of cartilage catabolic (Adamts4, Mmp13) and inflammatory genes (Nos2, Il6) was significantly elevated in cartilage from 6-month-old hTTR-TG mice compared with WT mice as was the level of phospho-NF-κB p65. Intra-articular injection of aggregated TTR in WT mice increased synovitis and significantly increased expression of inflammatory genes in synovium. These findings are the first to show that TTR deposition increases disease severity in the murine DMM and aging model of OA.
Collapse
Affiliation(s)
- Tokio Matsuzaki
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCAUSA
| | - Yukio Akasaki
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCAUSA
| | - Merissa Olmer
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCAUSA
| | | | - Natalia Reixach
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCAUSA
| | - Joel N. Buxbaum
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCAUSA
| | - Martin K. Lotz
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCAUSA
| |
Collapse
|