1
|
Ali Assar MF, Abd El Gayed EM, Abd El-Hamid Ewis AS, Zaid AB, Abd Allah Mahmoud Fouda E. Biochemical study of ZNF76 rs10947540 and SCUBE3 rs1888822 single nucleotide polymorphisms in the Egyptian patients with systemic lupus Erythematosus. J Immunoassay Immunochem 2024; 45:415-431. [PMID: 38982741 DOI: 10.1080/15321819.2024.2371590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Systemic lupus erythematosus (SLE) is a common autoimmune disease marked by the formation of apoptotic debris and the presence of autoantibodies that target nuclear components. At this moment, the actual cause of SLE is uncertain. Genetic variables have been well proven to have a significant role in the propensity of SLE. This study aimed to investigate the effect of (ZNF76) rs (10947540) and (SCUBE) rs (1888822) gene polymorphism in patients with systemic lupus erythematosus. A case control study has been carried out at Medical Biochemistry & Molecular biology and Rheumatology unit of Internal Medicine Departments, Faculty of Medicine, Menoufia University, Egypt, for 1-year duration between 1 June 2022 and 1 June 2023. Sixty patients were females (75%) and twenty patients were males (25%). Their ages ranged from 19 to 53 years. Their disease durations ranged from 7 months to 20 years. The findings indicated that the TC genotype of the ZNF76 rs10947540 gene increases the risk of SLE by 2.274-fold, while the dominant TC + CC increases the risk by 2.472-fold, and the C allele increases the risk by 2.115-fold. Additionally, the results showed that the TT genotype of the SCUBE3 rs1888822 gene increases the risk of SLE by 3.702-fold, the dominant GT + TT increases the risk by 2.304-fold, and the T allele increases the risk by 2.089-fold, while the GT genotype increases the risk by 1.918-fold. The study revealed significant associations between the genotypes of these polymorphisms and certain clinical parameters in SLE patients. These findings highlight the potential genetic contributions to SLE susceptibility and its clinical manifestations, providing valuable insights for future research and potential personalized approaches to the management of this complex autoimmune disease.
Collapse
Affiliation(s)
- Mohamed Farag Ali Assar
- Department of Chemistry, Biochemistry Division, Faculty of Science, Menoufia University, Shibin al Kawm, Egypt
| | - Eman Masoud Abd El Gayed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Menoufia University, Shibin al Kawm, Egypt
| | | | - Ahmed B Zaid
- Departments of Clinical Pathology, National Liver Institute, Menoufia University, Shibin Elkom, Egypt
| | | |
Collapse
|
2
|
Dar AA, Kim DD, Gordon SM, Klinzing K, Rosen S, Guha I, Porter N, Ortega Y, Forsyth KS, Roof J, Fazelinia H, Spruce LA, Eisenlohr LC, Behrens EM, Oliver PM. c-Myc uses Cul4b to preserve genome integrity and promote antiviral CD8 + T cell immunity. Nat Commun 2023; 14:7098. [PMID: 37925424 PMCID: PMC10625626 DOI: 10.1038/s41467-023-42765-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023] Open
Abstract
During infection, virus-specific CD8+ T cells undergo rapid bursts of proliferation and differentiate into effector cells that kill virus-infected cells and reduce viral load. This rapid clonal expansion can put T cells at significant risk for replication-induced DNA damage. Here, we find that c-Myc links CD8+ T cell expansion to DNA damage response pathways though the E3 ubiquitin ligase, Cullin 4b (Cul4b). Following activation, c-Myc increases the levels of Cul4b and other members of the Cullin RING Ligase 4 (CRL4) complex. Despite expressing c-Myc at high levels, Cul4b-deficient CD8+ T cells do not expand and clear the Armstrong strain of lymphocytic choriomeningitis virus (LCMV) in vivo. Cul4b-deficient CD8+ T cells accrue DNA damage and succumb to proliferative catastrophe early after antigen encounter. Mechanistically, Cul4b knockout induces an accumulation of p21 and Cyclin E2, resulting in replication stress. Our data show that c-Myc supports cell proliferation by maintaining genome stability via Cul4b, thereby directly coupling these two interdependent pathways. These data clarify how CD8+ T cells use c-Myc and Cul4b to sustain their potential for extraordinary population expansion, longevity and antiviral responses.
Collapse
Affiliation(s)
- Asif A Dar
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Dale D Kim
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Scott M Gordon
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathleen Klinzing
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Siera Rosen
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ipsita Guha
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nadia Porter
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yohaniz Ortega
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Katherine S Forsyth
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jennifer Roof
- Division of Cell Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hossein Fazelinia
- Division of Cell Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lynn A Spruce
- Division of Cell Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laurence C Eisenlohr
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward M Behrens
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Paula M Oliver
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Macedo JMB, Silva AL, Pinto AC, Landeira LFL, Portari EA, Santos-Rebouças CB, Klumb EM. TP53 and p21 (CDKN1A) polymorphisms and the risk of systemic lupus erythematosus. Adv Rheumatol 2023; 63:43. [PMID: 37605254 DOI: 10.1186/s42358-023-00320-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND The p53 and p21 proteins are important regulators of cell cycle and apoptosis and may contribute to autoimmune diseases, such as systemic lupus erythematosus (SLE). As genetic polymorphisms may cause changes in protein levels and functions, we investigated associations of TP53 and p21 (CDKN1A) polymorphisms (p53 72 G > C-rs1042522; p53 PIN3-rs17878362; p21 31 C > A-rs1801270; p21 70 C > T-rs1059234) with the development of systemic lupus erythematosus (SLE) in a Southeastern Brazilian population. METHODS Genotyping of 353 female volunteers (cases, n = 145; controls, n = 208) was performed by polymerase chain reaction, restriction fragment length polymorphism and/or DNA sequencing. Associations between TP53 and p21 polymorphisms and SLE susceptibility and clinical manifestations of SLE patients were assessed by logistic regression analysis. RESULTS Protective effect was observed for the genotype combinations p53 PIN3 A1/A1-p21 31 C/A, in the total study population (OR 0.45), and p53 PIN3 A1/A2-p21 31 C/C, in non-white women (OR 0.28). In Whites, p53 72 C-containing (OR 3.06) and p53 PIN3 A2-containing (OR 6.93) genotypes were associated with SLE risk, and higher OR value was observed for the combined genotype p53 72 G/C-p53 PIN3 A1/A2 (OR 9.00). Further, p53 PIN3 A1/A2 genotype was associated with serositis (OR 2.82), while p53 PIN3 A2/A2 and p53 72 C/C genotypes were associated with neurological disorders (OR 4.69 and OR 3.34, respectively). CONCLUSIONS Our findings showed that the TP53 and p21 polymorphisms included in this study may have potential to emerge as SLE susceptibility markers for specific groups of patients. Significant interactions of the TP53 polymorphisms with serositis and neurological disorders were also observed in SLE patients.
Collapse
Affiliation(s)
| | - Amanda Lima Silva
- Department of Biochemistry, State University of Rio de Janeiro - UERJ, Rio de Janeiro, Brazil
| | - Amanda Chaves Pinto
- Department of Biochemistry, State University of Rio de Janeiro - UERJ, Rio de Janeiro, Brazil
| | | | - Elyzabeth Avvad Portari
- Department of Pathological Anatomy, State University of Rio de Janeiro - UERJ, Rio de Janeiro, Brazil
- Department of Pathology, Fernandes Figueira Institute - FIOCRUZ, Rio de Janeiro, Brazil
| | | | - Evandro Mendes Klumb
- Department of Rheumatology, Pedro Ernesto University Hospital, State University of Rio de Janeiro - UERJ, Boulevard 28 de Setembro, 87, Vila Isabel, Rio de Janeiro, RJ, CEP 20551-030, Brazil.
| |
Collapse
|
4
|
Sturmlechner I, Zhang C, Sine CC, van Deursen EJ, Jeganathan KB, Hamada N, Grasic J, Friedman D, Stutchman JT, Can I, Hamada M, Lim DY, Lee JH, Ordog T, Laberge RM, Shapiro V, Baker DJ, Li H, van Deursen JM. p21 produces a bioactive secretome that places stressed cells under immunosurveillance. Science 2021; 374:eabb3420. [PMID: 34709885 PMCID: PMC8985214 DOI: 10.1126/science.abb3420] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immune cells identify and destroy damaged cells to prevent them from causing cancer or other pathologies by mechanisms that remain poorly understood. Here, we report that the cell-cycle inhibitor p21 places cells under immunosurveillance to establish a biological timer mechanism that controls cell fate. p21 activates retinoblastoma protein (Rb)–dependent transcription at select gene promoters to generate a complex bioactive secretome, termed p21-activated secretory phenotype (PASP). The PASP includes the chemokine CXCL14, which promptly attracts macrophages. These macrophages disengage if cells normalize p21 within 4 days, but if p21 induction persists, they polarize toward an M1 phenotype and lymphocytes mount a cytotoxic T cell response to eliminate target cells, including preneoplastic cells. Thus, p21 concurrently induces proliferative arrest and immunosurveillance of cells under duress.
Collapse
Affiliation(s)
- Ines Sturmlechner
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Chance C. Sine
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Erik-Jan van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Karthik B. Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Naomi Hamada
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Jan Grasic
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - David Friedman
- Department of Immunology, Mayo Clinic, Rochester MN, United States
| | - Jeremy T. Stutchman
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Ismail Can
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester MN, United States
| | - Masakazu Hamada
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Do Young Lim
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Jeong-Heon Lee
- Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester MN, United States
| | - Tamas Ordog
- Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester MN, United States
| | - Remi-Martin Laberge
- Unity Biotechnology, 285 E Grand Ave., South San Francisco, California 94080, USA
| | - Virginia Shapiro
- Department of Immunology, Mayo Clinic, Rochester MN, United States
| | - Darren J. Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester MN, United States
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Jan M. van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester MN, United States
| |
Collapse
|
5
|
de Paiva CS, Trujillo-Vargas CM, Schaefer L, Yu Z, Britton RA, Pflugfelder SC. Differentially Expressed Gene Pathways in the Conjunctiva of Sjögren Syndrome Keratoconjunctivitis Sicca. Front Immunol 2021; 12:702755. [PMID: 34349764 PMCID: PMC8326832 DOI: 10.3389/fimmu.2021.702755] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Sjögren syndrome (SS) is an autoimmune condition that targets the salivary and lacrimal glands, with cardinal clinical signs of dry eye (keratoconjunctivitis sicca, KCS) and dry mouth. The conjunctiva of SS patients is often infiltrated by immune cells that participate in the induction and maintenance of local inflammation. The purpose of this study was to investigate immune-related molecular pathways activated in the conjunctiva of SS patients. Female SS patients (n=7) and controls (n=19) completed a series of oral, ocular surface exams. Symptom severity scores were evaluated using validated questionnaires (OSDI and SANDE). All patients fulfilled the ACR/EULAR criteria for SS and the criteria for KCS. Fluorescein and lissamine green dye staining evaluated tear-break-up time (TBUT), corneal and conjunctival disease, respectively. Impression cytology of the temporal bulbar conjunctiva was performed to collect cells lysed and subjected to gene expression analysis using the NanoString Immunology Panel. 53/594 differentially expressed genes (DEGs) were observed between SS and healthy controls; 49 DEGs were upregulated, and 4 were downregulated (TRAF5, TGFBI, KLRAP1, and CMKLRI). The top 10 DEGs in descending order were BST2, IFITM1, LAMP3, CXCL1, IL19, CFB, LY96, MX1, IL4R, CDKN1A. Twenty pathways had a global significance score greater or equal to 2. Spearman correlations showed that 29/49 upregulated DEGs correlated with either TBUT (inverse) or OSDI or conjunctival staining score (positive correlations). Venn diagrams identified that 26/29 DEGs correlated with TBUT, 5/26 DEGs correlated with OSDI, and 16/26 correlated with conjunctival staining scores. Five upregulated DEGs (CFB, CFI, IL1R1, IL2RG, IL4R) were uniquely negatively correlated with TBUT. These data indicate that the conjunctiva of SS patients exhibits a phenotype of immune activation, although some genes could be inhibitory. Some of the DEGs and pathways overlap with previous DEGs in salivary gland biopsies, but new DEGs were identified, and some of these correlated with symptoms and signs of dry eye. Our results indicate that gene analysis of conjunctiva imprints is a powerful tool to understand the pathogenesis of SS and develop new therapeutic targets.
Collapse
Affiliation(s)
- Cintia S. de Paiva
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Claudia M. Trujillo-Vargas
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
- Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
- Grupo de Inmunodeficiencias Primarias, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Laura Schaefer
- Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Zhiyuan Yu
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Robert A. Britton
- Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | | |
Collapse
|
6
|
Manolakou T, Verginis P, Boumpas DT. DNA Damage Response in the Adaptive Arm of the Immune System: Implications for Autoimmunity. Int J Mol Sci 2021; 22:5842. [PMID: 34072535 PMCID: PMC8198144 DOI: 10.3390/ijms22115842] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022] Open
Abstract
In complex environments, cells have developed molecular responses to confront threats against the genome and achieve the maintenance of genomic stability assuring the transfer of undamaged DNA to their progeny. DNA damage response (DDR) mechanisms may be activated upon genotoxic or environmental agents, such as cytotoxic drugs or ultraviolet (UV) light, and during physiological processes requiring DNA transactions, to restore DNA alterations that may cause cellular malfunction and affect viability. In addition to the DDR, multicellular organisms have evolved specialized immune cells to respond and defend against infections. Both adaptive and innate immune cells are subjected to DDR processes, either as a prerequisite to the immune response, or as a result of random endogenous and exogenous insults. Aberrant DDR activities have been extensively studied in the immune cells of the innate arm, but not in adaptive immune cells. Here, we discuss how the aberrant DDR may lead to autoimmunity, with emphasis on the adaptive immune cells and the potential of therapeutic targeting.
Collapse
Affiliation(s)
- Theodora Manolakou
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece;
- School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Panayotis Verginis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, 700 13 Heraklion, Greece;
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, University of Crete Medical School, 700 13 Heraklion, Greece
| | - Dimitrios T. Boumpas
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece;
- Joint Rheumatology Program, 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, 124 62 Athens, Greece
| |
Collapse
|
7
|
The ZNF76 rs10947540 polymorphism associated with systemic lupus erythematosus risk in Chinese populations. Sci Rep 2021; 11:5186. [PMID: 33664275 PMCID: PMC7933287 DOI: 10.1038/s41598-021-84236-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a typical autoimmune disease with a strong genetic disposition. Genetic studies have revealed that single-nucleotide polymorphisms (SNPs) in zinc finger protein (ZNF)-coding genes are associated with susceptibility to autoimmune diseases, including SLE. The objective of the current study was to evaluate the correlation between ZNF76 gene polymorphisms and SLE risk in Chinese populations. A total of 2801 individuals (1493 cases and 1308 controls) of Chinese Han origin were included in this two-stage genetic association study. The expression of ZNF76 was evaluated, and integrated bioinformatic analysis was also conducted. The results showed that 28 SNPs were associated with SLE susceptibility in the GWAS cohort, and the association of rs10947540 was successfully replicated in the independent replication cohort (Preplication = 1.60 × 10-2, OR 1.19, 95% CI 1.03-1.37). After meta-analysis, the association between rs10947540 and SLE was pronounced (Pmeta = 9.62 × 10-6, OR 1.29, 95% CI 1.15-1.44). Stratified analysis suggested that ZNF76 rs10947540 C carriers were more likely to develop relatively high levels of serum creatinine (Scr) than noncarriers (CC + CT vs. TT, p = 9.94 × 10-4). The bioinformatic analysis revealed that ZNF76 rs10947540 was annotated as an eQTL and that rs10947540 was correlated with decreased expression of ZNF76. Remarkably, significantly reduced expression of ZNF76 was confirmed by expression data from both our laboratory and an array-based expression database. Taken together, these results suggest that ZNF76 rs10947540 is a possible susceptibility factor associated with SLE susceptibility. The mechanism underlying the relationship between ZNF76 and SLE pathogenesis still requires further investigation.
Collapse
|
8
|
Seleznik GM, Reding T, Peter L, Gupta A, Steiner SG, Sonda S, Verbeke CS, Dejardin E, Khatkov I, Segerer S, Heikenwalder M, Graf R. Development of autoimmune pancreatitis is independent of CDKN1A/p21-mediated pancreatic inflammation. Gut 2018; 67:1663-1673. [PMID: 28774888 DOI: 10.1136/gutjnl-2016-313458] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 06/07/2017] [Accepted: 06/19/2017] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Chronic pancreatitis (CP) and autoimmune pancreatitis (AIP) are characterised by different inflammatory processes. If pancreatic inflammation is a prerequisite for autoimmunity is still unclear. AIP is considered mostly a T cell-mediated disease; however, in induction of CP, macrophages play a pivotal role. p21-a member of cyclin-dependent kinase inhibitors-can influence inflammatory processes, in particular can regulate T cell activation and promote macrophage development. We therefore examined the role of p21-mediated inflammation in AIP. DESIGN We intercrossed lymphotoxin (LT) overexpressing mice (Tg(Ela1-LTa,b))-a model to study AIP development-with p21-deficient mice. Furthermore, we characterised p21 expression in human AIP and non-AIP specimens. RESULTS p21 deficiency in LT mice (LTp21-/-) prevented early pancreatic injury and reduced inflammation. In acinar cells, diminished proliferation and abrogated activation of non-canonical nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) pathway was observed. In contrast, 12-month-old LT mice with and without p21 had similar inflammatory signatures and T-B cell infiltration. Interestingly, LT and LTp21-/- mice had comparable tertiary lymphoid organs (TLOs), autoantibodies and elevated IgG levels. However, acinar cell proliferation, acinar-to-ductal metaplasia and acinar non-canonical NF-κB pathway activation remained impaired in LTp21-/- pancreata. CONCLUSIONS Our findings indicate that p21 is crucial for pancreatic inflammation in LT-driven pancreatic injury. p21 is involved in early acinar secretion of inflammatory mediators that attract innate immune cells. However, p21 is not essential for humoral immune response, accountable for autoimmunity. Remarkably, p21 renders acinar cells less susceptible to proliferation and transdifferentiation. We therefore suggest that AIP can also develop independent of chronic inflammatory processes.
Collapse
Affiliation(s)
- Gitta M Seleznik
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Theresia Reding
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Lukas Peter
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Anurag Gupta
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Sabrina G Steiner
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Sabrina Sonda
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Caroline S Verbeke
- Department of Pathology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Research, University of Liège, Liège, Belgium
| | - Igor Khatkov
- Department of Medicine and Dentistry, Moscow State University, Moscow, Russia
| | - Stephan Segerer
- Division of Nephrology, University Hospital, Zurich, Switzerland.,Division of Nephrology, Dialysis and Transplantation, Kantonsspital Aarau, Aarau, Switzerland
| | - Mathias Heikenwalder
- School of Medicine, Institute of Virology, TUM-Helmholtz Zentrum Munich, Munich, Germany.,Department of Chronic Inflammation and Cancer, German Cancer Center (DKFZ), Heidelberg, Germany
| | - Rolf Graf
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Iguchi T, Aoki K, Ikawa T, Taoka M, Taya C, Yoshitani H, Toma-Hirano M, Koiwai O, Isobe T, Kawamoto H, Masai H, Miyatake S. BTB-ZF Protein Znf131 Regulates Cell Growth of Developing and Mature T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:982-93. [PMID: 26136427 DOI: 10.4049/jimmunol.1500602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/31/2015] [Indexed: 02/01/2023]
Abstract
Many members of the BTB-ZF family have been shown to play important roles in lymphocyte development and function. The role of zinc finger Znf131 (also known as Zbtb35) in T cell lineage was elucidated through the production of mice with floxed allele to disrupt at different stages of development. In this article, we present that Znf131 is critical for T cell development during double-negative to double-positive stage, with which significant cell expansion triggered by the pre-TCR signal is coupled. In mature T cells, Znf131 is required for the activation of effector genes, as well as robust proliferation induced upon TCR signal. One of the cyclin-dependent kinase inhibitors, p21(Cip1) encoded by cdkn1a gene, is one of the targets of Znf131. The regulation of T cell proliferation by Znf131 is in part attributed to its suppression on the expression of p21(Cip1).
Collapse
Affiliation(s)
- Tomohiro Iguchi
- Laboratory of Self Defense Gene Regulation, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda 278-8510, Japan
| | - Kazuhisa Aoki
- Laboratory of Self Defense Gene Regulation, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Tomokatsu Ikawa
- Young Chief Investigators Laboratory for Immune Regeneration, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Masato Taoka
- Laboratory of Biochemistry, Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Choji Taya
- Animal Research Division, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Hiroshi Yoshitani
- Laboratory of Self Defense Gene Regulation, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Makiko Toma-Hirano
- Department of Otolaryngology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Osamu Koiwai
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda 278-8510, Japan
| | - Toshiaki Isobe
- Laboratory of Biochemistry, Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Hiroshi Kawamoto
- Department of Immunology, Field of Regeneration Control, Institute of Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan; and
| | - Hisao Masai
- Genome Dynamics Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Shoichiro Miyatake
- Laboratory of Self Defense Gene Regulation, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| |
Collapse
|
10
|
Crampton SP, Morawski PA, Bolland S. Linking susceptibility genes and pathogenesis mechanisms using mouse models of systemic lupus erythematosus. Dis Model Mech 2015; 7:1033-46. [PMID: 25147296 PMCID: PMC4142724 DOI: 10.1242/dmm.016451] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Systemic lupus erythematosus (SLE) represents a challenging autoimmune disease from a clinical perspective because of its varied forms of presentation. Although broad-spectrum steroids remain the standard treatment for SLE, they have many side effects and only provide temporary relief from the symptoms of the disease. Thus, gaining a deeper understanding of the genetic traits and biological pathways that confer susceptibility to SLE will help in the design of more targeted and effective therapeutics. Both human genome-wide association studies (GWAS) and investigations using a variety of mouse models of SLE have been valuable for the identification of the genes and pathways involved in pathogenesis. In this Review, we link human susceptibility genes for SLE with biological pathways characterized in mouse models of lupus, and discuss how the mechanistic insights gained could advance drug discovery for the disease.
Collapse
Affiliation(s)
- Steve P Crampton
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Peter A Morawski
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Silvia Bolland
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
11
|
Xu Z, Morel L. Contribution of B-1a cells to systemic lupus erythematosus in the NZM2410 mouse model. Ann N Y Acad Sci 2015; 1362:215-23. [PMID: 25728381 DOI: 10.1111/nyas.12607] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease of complex etiology in which B cells play a central role. An expanded number of B-1a cells have been consistently associated with murine lupus, and more recently with human SLE. We have identified Cdkn2c, a gene that controls cell cycle progression, as a key regulator of B-1a cell numbers and have associated Cdkn2c deficiency with autoimmune pathology, including the production of autoantibodies and the skewing of CD4(+) T cells toward inflammatory effector functions. We review the genetic studies that have led to these findings, as well as the possible mechanisms by which B-1a cell expansion and Cdkn2c deficiency are related to SLE pathogenesis.
Collapse
Affiliation(s)
- Zhiwei Xu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
12
|
Daszkiewicz L, Vázquez-Mateo C, Rackov G, Ballesteros-Tato A, Weber K, Madrigal-Avilés A, Di Pilato M, Fotedar A, Fotedar R, Flores JM, Esteban M, Martínez-A C, Balomenos D. Distinct p21 requirements for regulating normal and self-reactive T cells through IFN-γ production. Sci Rep 2015; 5:7691. [PMID: 25573673 PMCID: PMC4287747 DOI: 10.1038/srep07691] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/01/2014] [Indexed: 12/30/2022] Open
Abstract
Self/non-self discrimination characterizes immunity and allows responses against pathogens but not self-antigens. Understanding the principles that govern this process is essential for designing autoimmunity treatments. p21 is thought to attenuate autoreactivity by limiting T cell expansion. Here, we provide direct evidence for a p21 role in controlling autoimmune T cell autoreactivity without affecting normal T cell responses. We studied C57BL/6, C57BL/6/lpr and MRL/lpr mice overexpressing p21 in T cells, and showed reduced autoreactivity and lymphadenopathy in C57BL/6/lpr, and reduced mortality in MRL/lpr mice. p21 inhibited effector/memory CD4(+) CD8(+) and CD4(-)CD8(-) lpr T cell accumulation without altering defective lpr apoptosis. This was mediated by a previously non-described p21 function in limiting T cell overactivation and overproduction of IFN-γ, a key lupus cytokine. p21 did not affect normal T cell responses, revealing differential p21 requirements for autoreactive and normal T cell activity regulation. The underlying concept of these findings suggests potential treatments for lupus and autoimmune lymphoproliferative syndrome, without compromising normal immunity.
Collapse
Affiliation(s)
- Lidia Daszkiewicz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Cristina Vázquez-Mateo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Gorjana Rackov
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - André Ballesteros-Tato
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Kathrin Weber
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Adrián Madrigal-Avilés
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Mauro Di Pilato
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Arun Fotedar
- Cancer Cell Biology Program, Sidney Kimmel Cancer Center, San Diego, CA, USA
| | - Rati Fotedar
- Sanford-Burnham Medical Research Institute, San Diego, CA, USA
| | - Juana M Flores
- Animal Biology Department, School of Veterinary Medicine, Universidad Complutense, Madrid, Spain
| | - Mariano Esteban
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Carlos Martínez-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Dimitrios Balomenos
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| |
Collapse
|
13
|
Wells AD, Morawski PA. New roles for cyclin-dependent kinases in T cell biology: linking cell division and differentiation. Nat Rev Immunol 2014; 14:261-70. [PMID: 24603166 PMCID: PMC10114933 DOI: 10.1038/nri3625] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The proliferation of a few antigen-reactive lymphocytes into a large population of effector cells is a fundamental property of adaptive immunity. The cell division that fuels this process is driven by signals from antigen, co-stimulatory molecules and growth factor receptors, and is controlled by the cyclin-dependent kinase (CDK) cascade. In this Opinion article, we discuss how the CDK cascade provides one potential link between cell division and differentiation through the phosphorylation of immunologically relevant transcription factors, and how components of this pathway might ultimately participate in the decision between tolerance and immunity.
Collapse
Affiliation(s)
- Andrew D Wells
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, USA; and The Children's Hospital of Philadelphia Research Institute, Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA
| | - Peter A Morawski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, USA; and The Children's Hospital of Philadelphia Research Institute, Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
14
|
|
15
|
Rybakin V, Gascoigne NRJ. Negative selection assay based on stimulation of T cell receptor transgenic thymocytes with peptide-MHC tetramers. PLoS One 2012; 7:e43191. [PMID: 22900100 PMCID: PMC3416795 DOI: 10.1371/journal.pone.0043191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 07/20/2012] [Indexed: 02/04/2023] Open
Abstract
Thymocyte negative selection is a requirement for the development of self tolerance. Although it is possible to assay the induction of cell death in thymocytes in vitro using antibody cross-linking, this stimulus is much stronger than the normal range of T cell receptor ligands that could be encountered during normal development. Signaling in thymocytes is finely balanced between positive and negative selection stimuli, where a negative selecting ligand can be only marginally higher affinity than a positive selecting ligand. We have therefore developed an assay for the induction of negative selection that can distinguish such cases, and that is amenable to high-throughput analysis. The assay is based on the induction of activated caspase 3 in thymocytes expressing a defined T cell receptor, after stimulation with MHC-peptide tetramers in vitro for 24 hours or less.
Collapse
Affiliation(s)
- Vasily Rybakin
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail: (VR); (NRJG)
| | - Nicholas R. J. Gascoigne
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail: (VR); (NRJG)
| |
Collapse
|
16
|
Fairfax BP, Makino S, Radhakrishnan J, Plant K, Leslie S, Dilthey A, Ellis P, Langford C, Vannberg FO, Knight JC. Genetics of gene expression in primary immune cells identifies cell type-specific master regulators and roles of HLA alleles. Nat Genet 2012; 44:502-10. [PMID: 22446964 PMCID: PMC3437404 DOI: 10.1038/ng.2205] [Citation(s) in RCA: 388] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 01/31/2012] [Indexed: 12/15/2022]
Abstract
Trans-acting genetic variants have a substantial, albeit poorly characterized, role in the heritable determination of gene expression. Using paired purified primary monocytes and B cells, we identify new predominantly cell type-specific cis and trans expression quantitative trait loci (eQTLs), including multi-locus trans associations to LYZ and KLF4 in monocytes and B cells, respectively. Additionally, we observe a B cell-specific trans association of rs11171739 at 12q13.2, a known autoimmune disease locus, with IP6K2 (P = 5.8 × 10(-15)), PRIC285 (P = 3.0 × 10(-10)) and an upstream region of CDKN1A (P = 2 × 10(-52)), suggesting roles for cell cycle regulation and peroxisome proliferator-activated receptor γ (PPARγ) signaling in autoimmune pathogenesis. We also find that specific human leukocyte antigen (HLA) alleles form trans associations with the expression of AOAH and ARHGAP24 in monocytes but not in B cells. In summary, we show that mapping gene expression in defined primary cell populations identifies new cell type-specific trans-regulated networks and provides insights into the genetic basis of disease susceptibility.
Collapse
Affiliation(s)
- Benjamin P Fairfax
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Hoijman E, Rocha-Viegas L, Kalko SG, Rubinstein N, Morales-Ruiz M, Joffé EBDK, Kordon EC, Pecci A. Glucocorticoid alternative effects on proliferating and differentiated mammary epithelium are associated to opposite regulation of cell-cycle inhibitor expression. J Cell Physiol 2012; 227:1721-30. [PMID: 21688264 DOI: 10.1002/jcp.22896] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glucocorticoids influence post-natal mammary gland development by sequentially controlling cell proliferation, differentiation, and apoptosis. In the mammary gland, it has been demonstrated that glucocorticoid treatment inhibits epithelial apoptosis in post-lactating glands. In this study, our first goal was to identify new glucocorticoid target genes that could be involved in generating this effect. Expression profiling, by microarray analysis, revealed that expression of several cell-cycle control genes was altered by dexamethasone (DEX) treatment after lactation. Importantly, it was determined that not only the exogenous synthetic hormone, but also the endogenous glucocorticoids regulated the expression of these genes. Particularly, we found that the expression of cell cycle inhibitors p21CIP1, p18INK4c, and Atm was differentially regulated by glucocorticoids through the successive stages of mammary gland development. In undifferentiated cells, DEX treatment induced their expression and reduced cell proliferation, while in differentiated cells this hormone repressed expression of those cell cycle inhibitors and promoted survival. Therefore, differentiation status determined the effect of glucocorticoids on mammary cell fate. Particularly, we have determined that p21CIP1 inhibition would mediate the activity of these hormones in differentiated mammary cells because over-expression of this protein blocked DEX-induced apoptosis protection. Together, our data suggest that the multiple roles played by glucocorticoids in mammary gland development and function might be at least partially due to the alternative roles that these hormones play on the expression of cell cycle regulators.
Collapse
|
18
|
|
19
|
Bai H, Chen K, Gao YX, Arzigian M, Xie YL, Malcosky C, Yang YG, Wu WS, Wang ZZ. Bcl-xL enhances single-cell survival and expansion of human embryonic stem cells without affecting self-renewal. Stem Cell Res 2011; 8:26-37. [PMID: 22099018 DOI: 10.1016/j.scr.2011.08.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 08/03/2011] [Accepted: 08/06/2011] [Indexed: 12/21/2022] Open
Abstract
Robust expansion and genetic manipulation of human embryonic stem cells (hESCs) and induced-pluripotent stem (iPS) cells are limited by poor cell survival after enzymatic dissociation into single cells. Although inhibition of apoptosis is implicated for the single-cell survival of hESCs, the protective role of attenuation of apoptosis in hESC survival has not been elucidated. Bcl-xL is one of several anti-apoptotic proteins, which are members of the Bcl-2 family of proteins. Using an inducible system, we ectopically expressed Bcl-xL gene in hESCs, and found a significant increase of hESC colonies in the single-cell suspension cultures. Overexpression of Bcl-xL in hESCs decreased apoptotic caspase-3(+) cells, suggesting attenuation of apoptosis in hESCs. Without altering the kinetics of pluripotent gene expression, the efficiency to generate embryoid bodies (EBs) in vitro and the formation of teratoma in vivo were significantly increased in Bcl-xL-overexpressing hESCs after single-cell dissociation. Interestingly, the number and size of hESC colonies from cluster cultures were not affected by Bcl-xL overexpression. Several genes of extracellular matrix and adhesion molecules were upregulated by Bcl-xL in hESCs without single-cell dissociation, suggesting that Bcl-xL regulates adhesion molecular expression independent of cell dissociation. In addition, the gene expressions of FAS and several TNF signaling mediators were downregulated by Bcl-xL. These data support a model in which Bcl-xL promotes cell survival and increases cloning efficiency of dissociated hESCs without altering hESC self-renewal by i) attenuation of apoptosis, and ii) upregulation of adhesion molecules to facilitate cell-cell or cell-matrix interactions.
Collapse
Affiliation(s)
- Hao Bai
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Xenobiotic exposure and autoimmune hepatitis. HEPATITIS RESEARCH AND TREATMENT 2010; 2010:248157. [PMID: 21253536 PMCID: PMC3021850 DOI: 10.1155/2010/248157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 11/09/2010] [Indexed: 02/08/2023]
Abstract
Although genetics contributes to the development of autoimmune diseases, it is clear that "environmental" factors are also required. These factors are thought to encompass exposure to certain drugs and environmental pollutants. This paper examines the mechanisms that normally maintain immune unresponsiveness in the liver and discusses how exposure to certain xenobiotics such as trichloroethylene may disrupt those mechanisms and promote autoimmune hepatitis.
Collapse
|
21
|
Santiuste I, Buelta L, Iglesias M, Genre F, Mazorra F, Izui S, Merino J, Merino R. B-cell overexpression of Bcl-2 cooperates with p21 deficiency for the induction of autoimmunity and lymphomas. J Autoimmun 2010; 35:316-24. [PMID: 20691570 DOI: 10.1016/j.jaut.2010.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 07/08/2010] [Accepted: 07/09/2010] [Indexed: 11/13/2022]
Abstract
Genetic abnormalities predisposing to autoimmunity generally act in a cooperative manner affecting one or several mechanisms regulating immunological tolerance. In addition, many of these genetic abnormalities are also involved in the development of lymphoproliferative diseases. In the present study, we have determined the possible cooperation between deficiencies in members of the Cip/Kip family of cell cycle regulators (p21(WAF1/Cip1) or p27(kip1)) and the overexpression of human Bcl-2 in B lymphocytes in the induction of autoimmune and lymphoproliferative diseases in non-autoimmune C57BL/6 (B6) mice. Unlike single mutant mice, B6.p21(-/-) mice transgenic for human Bcl-2 in B cells developed a lethal autoimmune syndrome characterized by the production of autoantibodies, the prominent expansion of memory B and CD4(+) T cells and the development of severe glomerular lesions resembling IgA nephropathy. Furthermore, these mice presented a high incidence of B-cell lymphoproliferative disorders. Such genetic cooperation in the induction of autoimmunity was not observed in B6.p27(-/-) mice transgenic for human Bcl-2 in B cells. Altogether, what we have demonstrated here is the existence of preferential interactions among particular regulators of the G(1)/S transition of the cell cycle and B-cell survival in the induction of systemic autoimmune and lymphoproliferative diseases.
Collapse
Affiliation(s)
- Inés Santiuste
- Departmento de Biología Molecular, Universidad de Cantabria-Instituto de Formación e Investigación Marqués de Valdecilla, Santander, Spain
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Jacob N, Yang H, Pricop L, Liu Y, Gao X, Zheng SG, Wang J, Gao HX, Putterman C, Koss MN, Stohl W, Jacob CO. Accelerated pathological and clinical nephritis in systemic lupus erythematosus-prone New Zealand Mixed 2328 mice doubly deficient in TNF receptor 1 and TNF receptor 2 via a Th17-associated pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:2532-41. [PMID: 19201910 PMCID: PMC2790862 DOI: 10.4049/jimmunol.0802948] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
TNF-alpha has both proinflammatory and immunoregulatory functions. Whereas a protective role for TNF administration in systemic lupus erythematosus (SLE)-prone (New Zealand Black x New Zealand White)F(1) mice has been established, it remains uncertain whether this effect segregates at the individual TNFR. We generated SLE-prone New Zealand Mixed 2328 mice genetically deficient in TNFR1, in TNFR2, or in both receptors. Doubly-deficient mice developed accelerated pathological and clinical nephritis with elevated levels of circulating IgG anti-dsDNA autoantibodies and increased numbers of CD4(+) T lymphocytes, especially activated memory (CD44(high)CD62L(low)) CD4(+) T cells. We show that these cells expressed a Th17 gene profile, were positive for IL-17 intracellular staining by FACS, and produced exogenous IL-17 in culture. In contrast, immunological, pathological, and clinical profiles of mice deficient in either TNFR alone did not differ from those in each other or from those in wild-type controls. Thus, total ablation of TNF-alpha-mediated signaling was highly deleterious to the host in the New Zealand Mixed 2328 SLE model. These observations may have profound ramifications for the use of TNF and TNFR antagonists in human SLE and related autoimmune disorders, as well as demonstrate, for the first time, the association of the Th17 pathway with an animal model of SLE.
Collapse
MESH Headings
- Animals
- Antibodies, Antinuclear/blood
- Antibodies, Antinuclear/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Enzyme-Linked Immunosorbent Assay
- Female
- Flow Cytometry
- Fluorescent Antibody Technique
- Gene Expression Profiling
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Lupus Nephritis/genetics
- Lupus Nephritis/immunology
- Lupus Nephritis/pathology
- Mice
- Mice, Knockout
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type II/deficiency
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Noam Jacob
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
- Division of Gastrointestinal and Liver Diseases, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
| | - Haitao Yang
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
- Division of Rheumatology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
| | - Luminita Pricop
- Department of Medicine, Hospital for Special Surgery, Weill Medical College of Cornell University, New York, NY 10021
| | - Yi Liu
- Department of Medicine, Hospital for Special Surgery, Weill Medical College of Cornell University, New York, NY 10021
| | - Xiaoni Gao
- Department of Medicine, Hospital for Special Surgery, Weill Medical College of Cornell University, New York, NY 10021
| | - Song Guo Zheng
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
- Division of Rheumatology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
| | - Juhua Wang
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
- Division of Rheumatology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
| | - Hua-Xin Gao
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Chaim Putterman
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Michael N. Koss
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
| | - William Stohl
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
- Division of Rheumatology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
| | - Chaim O. Jacob
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
- Division of Gastrointestinal and Liver Diseases, University of Southern California Keck School of Medicine, Los Angeles, CA 90033
| |
Collapse
|
23
|
Heidari Y, Fossati-Jimack L, Carlucci F, Walport MJ, Cook HT, Botto M. A lupus-susceptibility C57BL/6 locus on chromosome 3 (Sle18) contributes to autoantibody production in 129 mice. Genes Immun 2008; 10:47-55. [PMID: 18843275 DOI: 10.1038/gene.2008.78] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
24
|
Zhu B, Symonds ALJ, Martin JE, Kioussis D, Wraith DC, Li S, Wang P. Early growth response gene 2 (Egr-2) controls the self-tolerance of T cells and prevents the development of lupuslike autoimmune disease. J Exp Med 2008; 205:2295-307. [PMID: 18779345 PMCID: PMC2556781 DOI: 10.1084/jem.20080187] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 08/07/2008] [Indexed: 11/08/2022] Open
Abstract
Maintaining tolerance of T cells to self-antigens is essential to avoid autoimmune disease. How self-reactive T cells are kept functionally inactive is, however, unknown. In this study, we show that early growth response gene 2 (Egr-2), a zinc-finger transcription factor, is expressed in CD44(high) T cells and controls their proliferation and activation. In the absence of Egr-2, CD44(high), but not CD44(low) T cells, are hyperreactive and hyperproliferative in vivo. The accumulation of activated CD4(+)CD44(high) T cells leads to the development of a late onset lupuslike autoimmune disease characterized by the accumulation of interferon (IFN)-gamma and interleukin (IL)-17-producing CD4(+) T cells, loss of tolerance to nuclear antigens, massive infiltration of T cells into multiple organs and glomerulonephritis. We found that the expression of cyclin-dependent kinase inhibitor p21cip1 was impaired in Egr-2-deficient T cells, whereas the expression of IFN-gamma and IL-17 in response to T cell receptor ligation was significantly increased, suggesting that Egr-2 activates the expression of genes involved in the negative regulation of T cell proliferation and inflammation. These results demonstrate that Egr-2 is an intrinsic regulator of effector T cells and controls the expansion of self-reactive T cells and development of autoimmune disease.
Collapse
Affiliation(s)
- Bo Zhu
- Institute of Cell and Molecular Science, Barts and London School of Medicine and Dentistry, University of London, London E1 2AT, England, UK
| | | | | | | | | | | | | |
Collapse
|
25
|
Bosque A, Aguiló JI, del Rey M, Paz-Artal E, Allende LM, Naval J, Anel A. Cell cycle regulation by FasL and Apo2L/TRAIL in human T-cell blasts. Implications for autoimmune lymphoproliferative syndromes. J Leukoc Biol 2008; 84:488-98. [PMID: 18483205 DOI: 10.1189/jlb.0108043] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The Fas-FasL pathway plays an important role in the homeostasis of mature lymphocytes, with defects causing autoimmune lymphoproliferative syndromes (ALPS). Human T-cell blasts are not sensitive to FasL or Apo2L/TRAIL-induced apoptosis unless they get reactivated, but either of those ligands inhibits their growth in the absence of cell death induction due to a cell cycle arrest in S-G2/M. In the present work, we have studied the mechanism(s) by which FasL or Apo2L/TRAIL regulate T-cell blast cell cycle in healthy donors and in two types of ALPS patients. Our data indicate that in human CD8+ T-cell blasts, Fas ligation, and especially Apo2L/TRAIL induce the p53-dependent decrease in cyclin-B1 levels. However, the induction of the negative cell cycle regulator p21WAF1 by FasL or Apo2L/TRAIL in either CD4+ or CD8+ T-cell blasts seems to be the main regulatory mechanism. This mechanism is dependent on caspase activation and on H2O2 generation. The increase in p21 levels by FasL or Apo2L/TRAIL is concomitant with p53 increases only in CD8+ T-cell blasts, with p21 levels maintained high for longer times than p53 levels. In CD4+ T-cell blasts p21 levels are controlled through a transient and p53-independent mechanism. The present results suggest that the etiology of ALP syndromes could be related not only to defects in apoptosis induction, but also in cell cycle regulation.
Collapse
Affiliation(s)
- Alberto Bosque
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza E-50009, Spain
| | | | | | | | | | | | | |
Collapse
|
26
|
Tarasenko T, Kole HK, Bolland S. A Lupus-Suppressor BALB/c Locus Restricts IgG2 Autoantibodies without Altering Intrinsic B Cell-Tolerance Mechanisms. THE JOURNAL OF IMMUNOLOGY 2008; 180:3807-14. [DOI: 10.4049/jimmunol.180.6.3807] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
27
|
Welling TH, Lu G, Csencsits K, Wood SC, Jarvinen L, Bishop DK. Regulation of alloimmune Th1 responses by the cyclin-dependent kinase inhibitor p21 following transplantation. Surgery 2008; 143:394-403. [PMID: 18291261 PMCID: PMC2423232 DOI: 10.1016/j.surg.2007.09.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Revised: 08/23/2007] [Accepted: 09/06/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND The cyclin-dependent kinase (cdk) inhibitor p21 inhibits cellular proliferation of many cell types, including T cells. Autoimmune models, however, have yielded conflicting results regarding the role of cdk inhibitors and T-cell function. The role of p21 in T-cell function after transplantation has not been investigated directly. We hypothesized that p21 plays an important role in alloantigen-driven responses in vitro in mixed lymphocyte cultures (MLC) and in vivo using the heterotopic murine cardiac allograft model. METHODS Wild type (WT) and p21-deficient (p21-/-) mice were used as recipients, and the effects of p21 overexpression were assessed by transplanting p21 adenoviral-transfected cardiac allografts. Enzyme-linked immunospot (ELISPOT) and 3H-thymidine incorporation were used to evaluate for T-cell priming and proliferation in vitro, whereas graft histology was evaluated for rejection. RESULTS When stimulated with alloantigens in vitro, splenocytes from p21-/- mice mounted enhanced proliferative responses and decreased Th2 responses relative to their WT counterparts. No differences in Th1 responses were noted when p21-/- cells were stimulated with alloantigens in vitro; however, after cardiac transplantation, Th1 responses were enhanced in p21-/- recipients relative to WT mice. This enhanced in vivo Th1 response was associated with exacerbated graft rejection in p21-/- recipients. Interestingly, p21 transfection of WT allografts inhibited graft rejection and Th1 priming. CONCLUSIONS p21 controls the intensity of the immune response posttransplantation, with overexpression inhibiting allograft rejection. Our data demonstrate that p21 controls T-cell priming and suggest that p21 and other cdk inhibitors may serve as potential targets for therapeutic manipulation of alloimmune responses.
Collapse
Affiliation(s)
- Theodore H Welling
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, Mich, USA.
| | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Santiago-Raber ML, Haraldsson MK, Theofilopoulos AN, Kono DH. Characterization of reciprocal Lmb1-4 interval MRL-Faslpr and C57BL/6-Faslpr congenic mice reveals significant effects from Lmb3. THE JOURNAL OF IMMUNOLOGY 2007; 178:8195-202. [PMID: 17548658 DOI: 10.4049/jimmunol.178.12.8195] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Susceptibility to severe lupus in MRL-Fas(lpr) mice requires not only the lpr mutation but also other predisposing genes. Using (MRL-Fas(lpr) x B6-Fas(lpr))F2 (where B6 represents C57BL/6) intercrosses that utilize the highly susceptible MRL and poorly susceptible B6 backgrounds, we previously mapped CFA-enhanced systemic lupus-like autoimmunity to four loci, named Lmb1-4, on chromosomes 4, 5, 7, and 10. In the current study, we generated and analyzed reciprocal interval congenic mice for susceptibility to CFA-enhanced autoimmunity at all four Lmb loci. Although all loci had at least a slight effect on lymphoproliferation, only Lmb3 demonstrated a major effect on lymphoproliferation and anti-chromatin Ab levels. Further characterization of Lmb3, primarily by comparing MRL-Fas(lpr) with MRL.B6-Lmb3 Fas(lpr) congenic mice, revealed that it also played a significant role in spontaneous lupus, modifying lymphoproliferation, IgG and autoantibody levels, kidney disease, and survival. The less susceptible B6 Lmb3 locus was associated with a marked reduction in numbers of CD4(+) and double-negative (CD4(-)CD8(-)) T cells, particularly in lymph nodes, as well as reduced T cell proliferation and enhanced T cell apoptosis, both in vivo and in vitro. IFN-gamma-producing CD4(+) T cells were also reduced in MRL.B6-Lmb3 Fas(lpr) mice. Further mapping using subinterval congenic mice placed Lmb3 in the telomeric portion of chromosome 7. Thus, Lmb3, primarily through its effects on CD4(+) and double-negative T cells, appears to be a highly penetrant lupus-modifying locus. Identification of the underlying genetic alteration responsible for this quantitative trait locus should provide new insights into lupus-modifying genes.
Collapse
|
30
|
Cauvi DM, Cauvi G, Pollard KM. Reduced expression of decay-accelerating factor 1 on CD4+ T cells in murine systemic autoimmune disease. ACTA ACUST UNITED AC 2007; 56:1934-44. [PMID: 17530719 DOI: 10.1002/art.22639] [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] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Deficiency of decay-accelerating factor 1 (termed Daf1 in mice) has been shown to exacerbate autoimmunity, and recent studies have suggested that this may be explained by Daf1 acting as a regulator of T cell immunity. The aim of this study was to determine whether Daf1 expression on T cells is modulated during development of autoimmunity in mice. METHODS To test this hypothesis, we examined Daf1 levels in NZB, DBA/2, and B10.S mice before and after induction of murine mercury-induced autoimmunity (mHgIA). Daf1 was measured by real-time polymerase chain reaction and flow cytometry, and levels of Daf1 were correlated with markers of lymphocyte activation and cytokine production. RESULTS Autoimmune-prone NZB mice had low endogenous levels of Daf1 irrespective of the induction of mHgIA. Induction of autoimmunity reduced Daf1 expression in mHgIA-sensitive B10.S mice, particularly on activated/memory (CD44(high)) CD4+ T cells that accumulate as a result of exposure to mercury. Murine mercury-induced autoimmunity-resistant DBA/2 mice, which fail to accumulate CD44(high) T cells, showed no change in Daf1 expression. Modulation of Daf1 expression was found to require CD4+ T cell costimulation, since B10.S mice deficient in CD28 were unable to down-regulate Daf1 or accumulate activated/memory CD4+ T cells. In B10.S mice exposed to mercury, the production of interleukin-4 (IL-4), but not that of IL-2 or interferon-gamma, in the spleen was associated with CD44(high),Daf1(low),CD4+ T cells. CONCLUSION These findings demonstrate that reduction of Daf1 expression is closely associated with CD4+ T cell activation and the accumulation of CD44(high)(activated/memory),CD4+ T cells in both spontaneous and induced systemic autoimmune disease.
Collapse
Affiliation(s)
- David M Cauvi
- W. M. Keck Autoimmune Disease Center, The Scripps Research Institute, La Jolla, California, USA
| | | | | |
Collapse
|
31
|
Wells AD. Cyclin-dependent kinases: molecular switches controlling anergy and potential therapeutic targets for tolerance. Semin Immunol 2007; 19:173-9. [PMID: 17383195 PMCID: PMC1949497 DOI: 10.1016/j.smim.2007.02.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Accepted: 02/16/2007] [Indexed: 11/18/2022]
Abstract
A large body of research has established the importance of costimulatory signals and proliferation for the generation of productive T cell immune responses. While costimulation and cell cycle progression are each individually necessary for CD4+ effector T cell differentiation, it has become clear that neither of these processes alone is sufficient to avoid anergy. This review outlines the links between T cell differentiation, tolerance, and the cell cycle, and highlights recent work that has implicated cyclin-dependent kinases as important regulators and potential targets for modulation of T cell immunity and tolerance.
Collapse
Affiliation(s)
- Andrew D Wells
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Joseph Stokes, Jr. Research Institute, The Children's Hospital of Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Arias CF, Ballesteros-Tato A, García MI, Martín-Caballero J, Flores JM, Martínez-A C, Balomenos D. p21CIP1/WAF1 Controls Proliferation of Activated/Memory T Cells and Affects Homeostasis and Memory T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2007; 178:2296-306. [PMID: 17277135 DOI: 10.4049/jimmunol.178.4.2296] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Development of autoantibodies and lupus-like autoimmunity by 129/Sv x C57BL/6 p21(-/-) mice has established that cell cycle deregulation is one the defective pathways leading to break of tolerance. Memory T cell accumulation is thought to be related to tolerance loss in murine lupus models. We studied T cell memory responses in C57BL/6 p21(-/-) mice that develop lupus-like disease manifestations. p21 did not affect primary proliferation of naive T cells, and was required for cycling control, but not for apoptosis of activated/memory T cells. When we induced apoptosis by secondary TCR challenge, surviving memory T cells depended on p21 for proliferation control. Under conditions of secondary T cell stimulation that did not cause apoptosis, p21 was also needed for regulation of activated/memory T cell expansion. The requirement for p21 in the control of T cell proliferation of activated/memory T cells suggests that in addition to apoptosis, cycling regulation by p21 constitutes a new pathway for T cell homeostasis. Concurring with this view, we found accumulation in p21(-/-) mice of memory CD4(+) T cells that showed increased proliferative potential after TCR stimulation. Furthermore, OVA immunization of p21(-/-) mice generated hyperresponsive OVA-specific T cells. Overall, the data show that p21 controls the proliferation of only activated/memory T cells, and suggest that p21 forms part of the memory T cell homeostasis mechanism, contributing to maintenance of tolerance.
Collapse
Affiliation(s)
- Cristina F Arias
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Cientificas, Campus de Cantoblanco, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
33
|
Unsinger J, Herndon JM, Davis CG, Muenzer JT, Hotchkiss RS, Ferguson TA. The role of TCR engagement and activation-induced cell death in sepsis-induced T cell apoptosis. THE JOURNAL OF IMMUNOLOGY 2007; 177:7968-73. [PMID: 17114469 DOI: 10.4049/jimmunol.177.11.7968] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Sepsis induces extensive apoptosis in T and B cells suggesting that the loss of immune effector cells could be one explanation for the profound immunosuppression observed in this disorder. Unfortunately, the mechanisms responsible for lymphocyte apoptosis in sepsis remain unknown. In T cells, apoptosis can occur through activation-induced cell death (AICD) in which engagement of the Ag receptors by cognate Ag or polyclonal activators such as bacteria-derived superantigens induces activation, proliferation, and apoptosis. We examined whether proliferation and AICD are necessary for apoptotic cell death in sepsis using normal and TCR transgenic mice. Results show that although sepsis resulted in activation of a small percentage of T cells, no proliferation was detected during the first 48 h following onset, a time when extensive apoptosis is observed. We also observed that T cells do not enter the cell cycle, and stimulation via the TCR in TCR transgenic animals does not enhance or decrease cell death in sepsis. Interestingly, T cells recovered from septic mice retained their ability to proliferate and synthesize cytokines albeit at reduced levels. With the exception of IL-10, which was increased in lymphocytes from mice with sepsis, sepsis caused a decrease in the production of both proinflammatory and anti-inflammatory cytokines. We conclude that lymphocyte apoptosis in sepsis does not require proliferation, TCR engagement, or AICD. Thus the immunosuppression observed in sepsis cannot be the result of T cell deletion via the TCR.
Collapse
Affiliation(s)
- Jacqueline Unsinger
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
34
|
Griffin SV, Olivier JP, Pippin JW, Roberts JM, Shankland SJ. Cyclin I Protects Podocytes from Apoptosis. J Biol Chem 2006; 281:28048-57. [PMID: 16847066 DOI: 10.1074/jbc.m513336200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The limited regenerative capacity of the glomerular podocyte following injury underlies the development of glomerulosclerosis and progressive renal failure in a diverse range of kidney diseases. We discovered that, in the kidney, cyclin I is uniquely expressed in the glomerular podocyte, and have constructed cyclin I knock-out mice to explore the biological function of cyclin I in these cells. Cyclin I knock-out (-/-) podocytes showed an increased susceptibility to apoptosis both in vitro and in vivo. Following induction of experimental glomerulonephritis, podocyte apoptosis was increased 4-fold in the cyclin I -/- mice, which was associated with dramatically decreased renal function. Our previous data showed that the Cdk inhibitor p21(Cip1/Waf1) protects podocytes from certain apoptotic stimuli. In cultured cyclin I -/- podocytes, the level of p21(Cip1/Waf1) was lower at base line, had a shorter half-life, and declined more rapidly in response to apoptotic stimuli than in wild-type cells. Enforced expression of p21(Cip1/Waf1) reversed the susceptibility of cyclin I -/- podocytes to apoptosis. Cyclin I protects podocytes from apoptosis, and we provide preliminary data to suggest that this is mediated by stabilization of p21(Cip1/Waf1).
Collapse
Affiliation(s)
- Siân V Griffin
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, 98195, USA
| | | | | | | | | |
Collapse
|
35
|
Scatizzi JC, Hutcheson J, Bickel E, Woods JM, Klosowska K, Moore TL, Haines GK, Perlman H. p21Cip1 is required for the development of monocytes and their response to serum transfer-induced arthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1531-41. [PMID: 16651620 PMCID: PMC1606593 DOI: 10.2353/ajpath.2006.050555] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
One of the central functions of cyclin-dependent kinase inhibitors, such as p21, p27, or p16, is to prevent entry into the cell cycle. However, the question remains as to whether they have other functions in the cell. We previously demonstrated that overexpression of p21 in fibroblasts isolated from patients with rheumatoid arthritis decreases the production of pro-inflammatory molecules. Overexpression of p21 has been also shown to reduce the development of experimental arthritis in mice and rats. To explore the role of endogenous p21 in the development of arthritis, we induced arthritis in p21(-/-) mice using the K/BxN serum transfer model of arthritis. Mice deficient in p21 were more resistant to serum transfer-induced arthritis (K/BxN) than wild-type (wt) control mice. Fewer macrophages were detected in p21(-/-) as compared to wt joints following transfer of K/BxN serum. Chemotaxis assays of bone marrow-derived macrophages from p21(-/-) and wt mice revealed no difference in migration. However, there was a substantial decrease in inflammatory monocytes circulating in peripheral blood and in monocyte precursors in bone marrow of p21(-/-) mice as compared to wt mice. Adoptive transfer of wt bone marrow-derived macrophages into p21(-/-) mice restored the sensitivity to serum transfer-induced arthritis. These data suggest a novel role for p21 in regulating the development and/or differentiation of monocytic populations that are crucial for the induction of inflammatory arthritis.
Collapse
Affiliation(s)
- John C Scatizzi
- Department of Molecular Microbiology and Immunology, Division of Rheumatology, School of Medicine, St. Louis University, St. Louis, Missouri 63104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Wang YH, Ito T, Wang YH, Homey B, Watanabe N, Martin R, Barnes CJ, McIntyre BW, Gilliet M, Kumar R, Yao Z, Liu YJ. Maintenance and polarization of human TH2 central memory T cells by thymic stromal lymphopoietin-activated dendritic cells. Immunity 2006; 24:827-838. [PMID: 16782037 DOI: 10.1016/j.immuni.2006.03.019] [Citation(s) in RCA: 247] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Revised: 02/17/2006] [Accepted: 03/14/2006] [Indexed: 11/19/2022]
Abstract
The identity of TH2 memory cells and the mechanism regulating their maintenance during allergic inflammation remain elusive. We report that circulated human CD4+ T cells expressing the prostaglandin D2 receptor (CRTH2) are TH2 central memory T cells, characterized by their phenotype, TH2 cytokine production, gene-expression profile, and the ability to respond to allergens. Only dendritic cells (DCs) activated by thymic stromal lymphopoietin (TSLP) can induce a robust expansion of CRTH2+CD4+ TH2 memory cells, while maintaining their central memory phenotype and TH2 commitments. CRTH2+CD4+ TH2 memory cells activated by TSLP-DCs undergo further TH2 polarization and express cystatin A, Charcot-Leydon crystal protein, and prostaglandin D2 synthase, implying their broader roles in allergic inflammation. Infiltrated CRTH2+CD4+ TH2 effector memory T cells in skin lesion of atopic dermatitis were associated with activated DCs, suggesting that TSLP-DCs play important roles not only in TH2 priming, but also in the maintenance and further polarization of TH2 central memory cells in allergic diseases.
Collapse
Affiliation(s)
- Yui-Hsi Wang
- Department of Immunology, Center of Cancer Immunology Research, Graduate School of Biomedical Science, Houston, Texas 77030
| | - Tomoki Ito
- Department of Immunology, Center of Cancer Immunology Research, Graduate School of Biomedical Science, Houston, Texas 77030
| | - Yi-Hong Wang
- Department of Immunology, Center of Cancer Immunology Research, Graduate School of Biomedical Science, Houston, Texas 77030
| | - Bernhard Homey
- Department of Dermatology, Heinrich-Heine-University, Düsseldorf 40225, Germany
| | - Norihiko Watanabe
- Department of Immunology, Center of Cancer Immunology Research, Graduate School of Biomedical Science, Houston, Texas 77030
| | | | - Christopher J Barnes
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Bradley W McIntyre
- Department of Immunology, Center of Cancer Immunology Research, Graduate School of Biomedical Science, Houston, Texas 77030
| | - Michel Gilliet
- Department of Immunology, Center of Cancer Immunology Research, Graduate School of Biomedical Science, Houston, Texas 77030
| | - Rakesh Kumar
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | | | - Yong-Jun Liu
- Department of Immunology, Center of Cancer Immunology Research, Graduate School of Biomedical Science, Houston, Texas 77030.
| |
Collapse
|
37
|
Bachmann MP, Bartsch H, Gross JK, Maier SM, Gross TF, Workman JL, James JA, Farris AD, Jung B, Franke C, Conrad K, Schmitz M, Büttner C, Buyon JP, Semsei I, Harley JB, Rieber EP. Autoimmunity as a result of escape from RNA surveillance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:1698-707. [PMID: 16849479 PMCID: PMC2206679 DOI: 10.4049/jimmunol.177.3.1698] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In previous studies, we detected a frame shift mutation in the gene encoding the autoantigen La of a patient with systemic lupus erythematosus. The mutant La mRNA contains a premature termination codon. mRNAs that prematurely terminate translation should be eliminated by RNA quality control mechanisms. As we find Abs specific for the mutant La form in approximately 30% of sera from anti-La-positive patients, we expected that mutant La mRNAs circumvent RNA control and the expression of mutant La protein could become harmful. Indeed, real-time PCR, immunostaining, and immunoblotting data of mice transgenic for the mutant La form show that mutant La mRNAs are not repressed in these animals and are translated to mutant La protein. In addition to the mutant La protein, we detected a minor portion of native human La in the mutant La-transgenic mice. Therefore, ribosomal frame shifting may allow the mutant La mRNA to escape from RNA control. Interestingly, expression of the mutant La mRNA results in a lupus-like disease in the experimental mice. Consequently, escape of mutant La mRNA from RNA control can have two effects: it 1) results in the expression of an immunogenic (neo)epitope, and 2) predisposes to autoimmunity.
Collapse
Affiliation(s)
- Michael P Bachmann
- Institute of Immunology, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Li WQ, Jiang Q, Aleem E, Kaldis P, Khaled AR, Durum SK. IL-7 promotes T cell proliferation through destabilization of p27Kip1. ACTA ACUST UNITED AC 2006; 203:573-82. [PMID: 16492801 PMCID: PMC2118250 DOI: 10.1084/jem.20051520] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Interleukin (IL)-7 is required for survival and homeostatic proliferation of T lymphocytes. The survival effect of IL-7 is primarily through regulation of Bcl-2 family members; however, the proliferative mechanism is unclear. It has not been determined whether the IL-7 receptor actually delivers a proliferative signal or whether, by promoting survival, proliferation results from signals other than the IL-7 receptor. We show that in an IL-7–dependent T cell line, cells protected from apoptosis nevertheless underwent cell cycle arrest after IL-7 withdrawal. This arrest was accompanied by up-regulation of the cyclin-dependent kinase inhibitor p27Kip1 through a posttranslational mechanism. Overexpression of p27Kip1 induced G1 arrest in the presence of IL-7, whereas knockdown of p27Kip1 by small interfering RNA promoted S phase entry after IL-7 withdrawal. CD4 or CD8 T cells transferred into IL-7–deficient hosts underwent G1 arrest, whereas 27Kip1-deficient T cells underwent proliferation. We observed that IL-7 withdrawal activated protein kinase C (PKC)θ and that inhibition of PKCθ with a pharmacological inhibitor completely blocked the rise of p27Kip1 and rescued cells from G1 arrest. The conventional pathway to breakdown of p27Kip1 is mediated by S phase kinase-associated protein 2; however, our evidence suggests that PKCθ acts via a distinct, unknown pathway inducing G1 arrest after IL-7 withdrawal from T cells. Hence, IL-7 maintains T cell proliferation through a novel pathway of p27Kip1 regulation.
Collapse
Affiliation(s)
- Wen Qing Li
- Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD 21702, USA
| | | | | | | | | | | |
Collapse
|
39
|
Thomas MJ, Agy MB, Proll SC, Paeper BW, Li Y, Jensen KL, Korth MJ, Katze MG. Functional gene analysis of individual response to challenge of SIVmac239 in M. mulatta PBMC culture. Virology 2006; 348:242-52. [PMID: 16430941 DOI: 10.1016/j.virol.2005.12.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Accepted: 12/06/2005] [Indexed: 11/27/2022]
Abstract
It has previously been shown in macaques that individual animals exhibit varying responses to challenge with the same strain of SIV. We attempted to elucidate these differences using functional genomics and correlate them to biological response. Unfractionated PBMC from three rhesus macaques were isolated, activated, and infected with SIVmac239. Interestingly, one of the three animals used for these experiments exhibited a completely unique response to infection relative to the other two. After repeated attempts to infect the PBMC from this animal, little or no infectivity was seen across the time points considered, and corresponding to this apparent lack of infection, few genes were seen to be differentially expressed when compared to mock-infected cells. For the remaining two animals, gene expression analysis showed that while they exhibited responses for the same groups of pathways, these responses included differences specific to the individual animal at the gene level. In instances where the patterns of differential gene expression differed between these animals, the genes being differentially expressed were associated with the same categories of biological process, mainly immune response and cell signaling. At the pathway level, these animals again exhibited similar responses that could be predicted based on the experimental conditions. Even in these expected results, the degree of response and the specific genes being regulated differed greatly from animal to animal. The differences in gene expression on an individual level have the potential to be used as markers in identification of animals suitable for lentiviral infection experiments. Our results highlight the importance of individual variation in response to viral challenge.
Collapse
Affiliation(s)
- Matthew J Thomas
- Department of Microbiology and Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Goulvestre C, Chéreau C, Nicco C, Mouthon L, Weill B, Batteux F. A mimic of p21WAF1/CIP1 ameliorates murine lupus. THE JOURNAL OF IMMUNOLOGY 2006; 175:6959-67. [PMID: 16272356 DOI: 10.4049/jimmunol.175.10.6959] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Systemic lupus erythematosus (SLE) is a progressive autoimmune disease characterized by the production of high levels of affinity-matured IgG autoantibodies to dsDNA and, possibly, visceral involvement. Pathogenic autoantibodies result from the activation and proliferation of autoreactive T and B lymphocytes stimulated by epitopes borne by nucleosomal histones. To inhibit the proliferation of autoreactive cells and abrogate the development of SLE, a novel tool, cell cycle inhibiting peptide therapy, was used. Thus, a peptidyl mimic of p21WAF1/CIP1 that inhibits the interaction between cyclin-dependent kinase 4 and type D cyclins abrogated the in vitro proliferative response of T cells to histones and T-independent and T-dependent proliferative responses of B cells. The WAF1/CIP1 mimic also abrogated the in vitro production of total and anti-dsDNA IgG Abs by B cells. Similarly, the p21WAF1/CIP1 construct inhibited the ex vivo T and B cell proliferative responses to histones and decreased the numbers of activated/memory B and T spleen cells. The alterations in the balance of spleen cell subsets resulted from proapoptotic effects of the p21WAF1/CIP1)construct on activated splenocytes. Finally, in vivo, four i.v. injections of the p21WAF1/CIP1 mimic were sufficient to inhibit the progression of the lupus-like syndrome in (NZB x NZW)F1 mice. The levels of anti-dsDNA IgG autoantibodies and the incidence and severity of renal involvement were lower in treated mice than in nontreated mice. Those observations open new avenues for the treatment of SLE and prompt us to evaluate the potential interest of peptidic therapy in human SLE.
Collapse
Affiliation(s)
- Claire Goulvestre
- Laboratoire d'Immunologie, Faculté de Médecine René Descartes et Hôpital Cochin, Université Paris 5, Paris, France
| | | | | | | | | | | |
Collapse
|
41
|
Bagavant H, Fu SM. New insights from murine lupus: disassociation of autoimmunity and end organ damage and the role of T cells. Curr Opin Rheumatol 2005; 17:523-8. [PMID: 16093828 DOI: 10.1097/01.bor.0000169361.23325.1e] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review summarizes current literature on genetic regulation of different phenotypes in systemic lupus erythematosus in context of end-organ disease. Recent findings conflicting with the current paradigm that loss of tolerance to chromatin is the critical step for end-organ injury are discussed. RECENT FINDINGS Systemic lupus erythematosus is a prototype immune complex disease with circulating autoantibodies to chromatin, histone proteins, Sm/La, and other nuclear and cytoplasmic proteins. Extensive studies have been carried out on the regulation of B-cell and autoantibody production in lupus mice. However, the hypothesis that autoantibodies are primary mediators of organ damage fails to explain the heterogenous presentation in patients. Studies in murine models of systemic lupus erythematosus clearly dissociate genetic control of autoantibody responses to classic lupus antigens and kidney disease. There is increasing evidence to support the role of autoreactive T cells and genetic control of end organ susceptibility. These studies suggest complex interactions between innate and adaptive immunity resulting in end-organ damage. This review focuses on autoimmune responses and renal involvement in spontaneous systemic lupus erythematosus using murine models of lupus nephritis. SUMMARY Studies in murine models demonstrate complex genetic interactions regulating spontaneous systemic lupus erythematosus. Although detection of serum autoantibodies is considered a hallmark for clinical diagnosis of systemic lupus erythematosus, recent evidence shows that autoantibodies to classic lupus antigens are neither required nor sufficient for end-organ damage. Thus, murine models provide new insights into the pathogenesis of systemic lupus erythematosus.
Collapse
Affiliation(s)
- Harini Bagavant
- Specialized Center of Research on Systemic Lupus Erythematosus, University of Virginia, Charlottesville, 22908, USA
| | | |
Collapse
|
42
|
Subramanian S, Yim YS, Liu K, Tus K, Zhou XJ, Wakeland EK. Epistatic suppression of systemic lupus erythematosus: fine mapping of Sles1 to less than 1 mb. THE JOURNAL OF IMMUNOLOGY 2005; 175:1062-72. [PMID: 16002707 DOI: 10.4049/jimmunol.175.2.1062] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sle is a susceptibility locus for systemic autoimmunity derived from the lupus-prone NZM2410 mouse. The New Zealand White-derived suppressive modifier Sles1 was identified as a specific modifier of Sle1 and prevents the development of IgG anti-chromatin autoantibodies mediated by Sle1 on the C57BL/6 (B6) background. Fine mapping of Sles1 with truncated congenic intervals localizes it to a approximately 956-kb segment of mouse chromosome 17. Sles1 completely abrogates the development of activated T and B cell populations in B6.Sle1. Despite this suppression of the Sle1-mediated cell surface activation phenotypes, B6.Sle1 Sles1 splenic B cells still exhibit intrinsic ERK phosphorylation. Classic genetic complementation tests using the nonautoimmmune 129/SvJ mouse suggests that this strain possesses a Sles1 allele complementary to that of New Zealand White, as evidenced by the lack of glomerulonephritis, splenomegaly, and antinuclear autoantibody production seen in (129 x B6.Sle1 Sles1)F(1)s. These findings localize and characterize the suppressive properties of Sles1 and implicate 129 as a useful strain for aiding in the identification of this elusive epistatic modifier gene.
Collapse
MESH Headings
- Animals
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cells, Cultured
- Epistasis, Genetic
- Female
- Genetic Complementation Test
- Immunophenotyping
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lymphocyte Activation/genetics
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Mice, Inbred NZB
- Mice, Inbred Strains
- Physical Chromosome Mapping/methods
- Spleen/immunology
- Spleen/metabolism
- Suppression, Genetic/immunology
- T-Lymphocytes/immunology
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- Srividya Subramanian
- Center for Immunology and Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
43
|
Vázquez N, Greenwell-Wild T, Marinos NJ, Swaim WD, Nares S, Ott DE, Schubert U, Henklein P, Orenstein JM, Sporn MB, Wahl SM. Human immunodeficiency virus type 1-induced macrophage gene expression includes the p21 gene, a target for viral regulation. J Virol 2005; 79:4479-91. [PMID: 15767448 PMCID: PMC1061522 DOI: 10.1128/jvi.79.7.4479-4491.2005] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In contrast to CD4+ T cells, human immunodeficiency virus type 1 (HIV-1)-infected macrophages typically resist cell death, support viral replication, and consequently, may facilitate HIV-1 transmission. To elucidate how the virus commandeers the macrophage's intracellular machinery for its benefit, we analyzed HIV-1-infected human macrophages for virus-induced gene transcription by using multiple parameters, including cDNA expression arrays. HIV-1 infection induced the transcriptional regulation of genes associated with host defense, signal transduction, apoptosis, and the cell cycle, among which the cyclin-dependent kinase inhibitor 1A (CDKN1A/p21) gene was the most prominent. p21 mRNA and protein expression followed a bimodal pattern which was initially evident during the early stages of infection, and maximum levels occurred concomitant with active HIV-1 replication. Mechanistically, viral protein R (Vpr) independently regulates p21 expression, consistent with the reduced viral replication and lack of p21 upregulation by a Vpr-negative virus. Moreover, the treatment of macrophages with p21 antisense oligonucleotides or small interfering RNAs reduced HIV-1 infection. In addition, the synthetic triterpenoid and peroxisome proliferator-activated receptor gamma ligand, 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO), which is known to influence p21 expression, suppressed viral replication. These data implicate p21 as a pivotal macrophage facilitator of the viral life cycle. Moreover, regulators of p21, such as CDDO, may provide an interventional approach to modulate HIV-1 replication.
Collapse
Affiliation(s)
- Nancy Vázquez
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wolfraim LA, Letterio JJ. Cutting edge: p27Kip1 deficiency reduces the requirement for CD28-mediated costimulation in naive CD8+ but not CD4+ T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2005; 174:2481-4. [PMID: 15728451 DOI: 10.4049/jimmunol.174.5.2481] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cell cycle re-entry of quiescent T cells is dependent upon cyclin-dependent kinase 2. Inhibition of cyclin-dependent kinase 2 by p27(Kip1) is believed to be the principal constraint on S-phase entry in T cells. We report that deficiency for p27(Kip1) has a more pronounced effect on the expansion of murine naive CD8(+) T cells and that this disparity is due to a reduced requirement for CD28-mediated costimulation in CD8(+) but not CD4(+) T cells lacking p27(Kip1). These data highlight a previously unappreciated difference in the way CD28 signaling is coupled to the core cell cycle machinery in these two T cell subsets.
Collapse
Affiliation(s)
- Lawrence A Wolfraim
- Laboratory of Cell Regulation and Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
45
|
Schmelz K, Wagner M, Dörken B, Tamm I. 5-Aza-2'-deoxycytidine induces p21WAF expression by demethylation of p73 leading to p53-independent apoptosis in myeloid leukemia. Int J Cancer 2005; 114:683-95. [PMID: 15609309 DOI: 10.1002/ijc.20797] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The DNA methylation inhibitor 5-Aza-2'-deoxycytidine (5-Aza-CdR) has significant therapeutic value for the treatment of patients with myelodysplastic syndrome (MDS), acute myeloid leukemia (AML) and chronic myeloid leukemia (CML). The demethylating effect of 5-Aza-CdR has been well characterized. In contrast, less is known about the molecular events downstream of the methylation inhibition. Here, 5-Aza-CdR induced apoptosis in AML cells (both p53 mutant and wild-type) but not in epithelial or normal PBMCs. Cell death was accompanied by activation of the mitochondrial apoptosis pathway, as shown by release of cytochrome c and AIF and loss of mitochondrial membrane potential (DeltaPsim). Activation of caspase-3 (but not -6 and -8) was detectable using Western blot analysis and measurement of caspase enzymatic activity. 5-Aza-CdR treatment resulted in the induction of p21, which correlated with the arrest of AML cells in the G1 cell cycle phase. Induction of p21 expression was independent of its promoter methylation status but mediated by 5-Aza-CdR-induced reexpression of the tumor-suppressor p73, a known upstream regulator of p21. The p73 promoter was hypermethylated in AML cell lines and in primary AML cells but not in epithelial cells, which were resistant toward 5-Aza-CdR. Therefore, 5-Aza-CdR-mediated specific killing of myeloid cells might be dependent on its ability to revert p73 promoter methylation and to reexpress p73 mRNA. In addition, exogenous expression of p73 rendered epithelial cells sensitive to apoptosis induced by 5-Aza-CdR or other cytostatic drugs. We therefore conclude that p73 is a relevant target for methylation-dependent efficacy of 5-Aza-CdR in AML cells.
Collapse
MESH Headings
- Apoptosis
- Azacitidine/analogs & derivatives
- Azacitidine/pharmacology
- Blotting, Western
- Caspase 3
- Caspases/metabolism
- Cell Cycle
- Cell Cycle Proteins/biosynthesis
- Cell Line, Tumor
- CpG Islands
- Cyclin-Dependent Kinase Inhibitor p21
- Cytochromes c/metabolism
- DNA Methylation
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Decitabine
- Epithelial Cells/cytology
- Epithelial Cells/metabolism
- Flow Cytometry
- G1 Phase
- Genes, Tumor Suppressor
- HL-60 Cells
- HeLa Cells
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myeloid, Acute/drug therapy
- Leukocytes, Mononuclear/metabolism
- Membrane Potentials
- Microscopy, Fluorescence
- Mitochondria/metabolism
- Myelodysplastic Syndromes/drug therapy
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Promoter Regions, Genetic
- RNA/chemistry
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sulfites/chemistry
- Time Factors
- Transfection
- Tumor Protein p73
- Tumor Suppressor Protein p53/metabolism
- Tumor Suppressor Proteins
- U937 Cells
- Up-Regulation
Collapse
Affiliation(s)
- Karin Schmelz
- Department of Hematology and Oncology, Universitätsmedizin Berlin, Charité, Campus Virchow, Berlin, Germany
| | | | | | | |
Collapse
|
46
|
Steinbach JP, Wolburg H, Klumpp A, Weller M. Hypoxia sensitizes human malignant glioma cells towards CD95L-induced cell death. J Neurochem 2005; 92:1340-9. [PMID: 15748153 DOI: 10.1111/j.1471-4159.2004.02957.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Death ligands such as CD95 ligand (CD95L) have limited activity against glioma cells under normoxic conditions. Hypoxia is a critical aspect of the microenvironment of gliomas in vivo. We investigated the effect of co-exposure to acute hypoxia and CD95 ligand in three human malignant glioma cell lines with different susceptibility to CD95L under normoxic conditions. Hypoxia sensitized all three cell lines towards CD95L-induced cell death. Co-exposure resulted in apoptotic changes in the early phase, with gradual conversion to secondary necrosis with increasing length of hypoxia. The mitochondrial injury induced by hypoxia was enhanced by co-treatment, and caspase cleavage became prominent. Inhibition of the epidermal growth factor receptor (EGFR), although sensitizing glioma cells to CD95L under normoxia, protects glioma cells from hypoxia by reducing energy consumption. However, the opposing effects of EGFR signalling on death induced by CD95L or hypoxia were neutralized by co-exposure to hypoxia and CD95L. Furthermore, inhibition of protein synthesis by cycloheximide also reduced glucose consumption and conferred protection from hypoxia, but did not modulate CD95L-induced cell death under hypoxic conditions. These results suggest that death ligands may be useful to target hypoxic tumour cells resistant to conventional therapies or to complement strategies aiming at the induction of tumour hypoxia.
Collapse
Affiliation(s)
- Joachim P Steinbach
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University of Tübingen, Medical School, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany.
| | | | | | | |
Collapse
|
47
|
Datta SK, Zhang L, Xu L. T-helper cell intrinsic defects in lupus that break peripheral tolerance to nuclear autoantigens. J Mol Med (Berl) 2005; 83:267-78. [PMID: 15630591 DOI: 10.1007/s00109-004-0624-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2004] [Accepted: 11/16/2004] [Indexed: 12/22/2022]
Abstract
Special populations of T helper cells drive B cells to produce IgG class switched, pathogenic autoantibodies in lupus. The major source of antigenic determinants (epitopes) that trigger interactions between lupus T and B cells is nucleosomes of apoptotic cells. These epitopes can be used for antigen-specific therapy of lupus. Secondly, the autoimmune T cells of lupus are sustained because they resist anergy and activation-induced programmed cell death by markedly upregulating cyclooxygenase (COX) 2 along with the antiapoptotic molecule c-FLIP. Only certain COX-2 inhibitors block pathogenic anti-DNA autoantibody production in lupus by causing death of autoimmune T helper cells. Hence COX-2 inhibitors may work independently of their ability to block the enzymatic function of COX-2, and structural peculiarities of these select inhibitors may lead to better drug discovery and design.
Collapse
Affiliation(s)
- Syamal K Datta
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 240 East Huron St., Chicago, IL 60611, USA.
| | | | | |
Collapse
|
48
|
Hadj-Slimane R, Chelbi-Alix MK, Tovey MG, Bobé P. An Essential Role for IFN-α in the Overexpression of Fas Ligand on MRL/lpr Lymphocytes and on Their Spontaneous Fas-Mediated Cytotoxic Potential. J Interferon Cytokine Res 2004; 24:717-28. [PMID: 15684739 DOI: 10.1089/jir.2004.24.717] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Lymphocytes from aged autoimmune MRL/lpr mice overexpress Fas ligand (FasL), and are cytotoxic against Fas+ target cells. This cytotoxic potential is only partly due to FasL, as wild-type MRL+/+ lymphocytes are not able to kill Fas+ targets after induction of FasL. In addition, serum levels of interferon-alpha (IFN-alpha) increase in parallel with the Fas-dependent cytotoxic potential of lymphocytes from MRL/lpr mice as they age. To understand the mechanisms underlying these observations, combined suppression subtractive hybridization (SSH) and RT-PCR were used to study differential gene expression in splenocytes from MRL/lpr mice compared with splenocytes from MRL+/+ mice. Twenty-two genes were upregulated transcriptionally in MRL/lpr splenocytes compared with their MRL+/+ counterparts. Furthermore, 9 of these genes were also upregulated after treatment of MRL/lpr splenocytes with IFN-alpha, and 4 were strongly downregulated. MRL/lpr lymphocytes were also found to be hyperresponsive to IFN-alpha. Thus, MRL/lpr lymphocytes overexpressed mRNA for the IFN-alpha receptor (IFNAR-1 and IFNAR-2) chains of the IFN-alpha/beta receptor and exhibited high endogenous levels of both Stat1 and phosphorylated Stat1 proteins. Lymphocytes from young MRL/lpr mice, with low Fas-dependent cytotoxic activity, were found to become highly cytotoxic against Fas+ targets after treatment with IFN-alpha. These data suggest that IFN-alpha plays an important role in the physiopathology of the systemic lupus erythematosus (SLE)-like syndrome that occurs in MRL/lpr mice.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Cytotoxicity, Immunologic/genetics
- DNA, Complementary/analysis
- DNA-Binding Proteins/metabolism
- Fas Ligand Protein
- Gene Expression Regulation
- Interferon-alpha/blood
- Interferon-alpha/pharmacology
- Interferon-alpha/physiology
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lymphocytes/drug effects
- Lymphocytes/immunology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred MRL lpr
- Nucleic Acid Hybridization
- Phosphorylation
- RNA, Messenger/analysis
- RNA, Messenger/metabolism
- Receptor, Interferon alpha-beta
- Receptors, Interferon/genetics
- Receptors, Interferon/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- STAT1 Transcription Factor
- Spleen/cytology
- Spleen/metabolism
- Trans-Activators/metabolism
- Transcription, Genetic/drug effects
- Transcriptional Activation
Collapse
Affiliation(s)
- Réda Hadj-Slimane
- Laboratoire d'Oncologie Virale, CNRS UPR 9045, Institut André Lwoff, Villejuif, France
| | | | | | | |
Collapse
|
49
|
Affiliation(s)
- M P Peppelenbosch
- Department of Gastroenterology and Hepatology, C2-330, Academic Medical Centre, Meibergdreef 9, NL-1105 AZ Amsterdam, the Netherlands
| | | |
Collapse
|
50
|
Abstract
Genomic instability is intrinsically linked to significant alterations in apoptosis control. Chromosomal and microsatellite instability can cause the inactivation of pro-apoptotic pathways. In addition, the inhibition of apoptosis itself can be permissive for the survival and ongoing division of cells that have failed to repair DNA double-strand breaks, experience telomere dysfunction or are in an abnormal polyploid state. Furthermore, DNA-repair proteins can regulate apoptosis. So, genomic instability and apoptosis are intimately linked phenomena, with important implications for the pathophysiology of cancer.
Collapse
Affiliation(s)
- Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, Nobels väg 13, SE-171 77 Stockholm, Sweden.
| | | |
Collapse
|