1
|
Huang L, Zhang C, Jiang A, Lin A, Zhu L, Mou W, Zeng D, Liu Z, Tang B, Zhang J, Cheng Q, Miao K, Wei T, Luo P. T-cell Senescence in the Tumor Microenvironment. Cancer Immunol Res 2025; 13:618-632. [PMID: 40232041 DOI: 10.1158/2326-6066.cir-24-0894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/07/2024] [Accepted: 01/24/2025] [Indexed: 04/16/2025]
Abstract
T-cell senescence occurs in the tumor microenvironment (TME) and influences cancer outcomes as well as the effectiveness of immunotherapies. The TME triggers this T-cell senescence via multiple pathways, including persistent stimulation with tumor-associated antigens, altered metabolic pathways, and activation of chronic inflammatory responses. Senescent T cells exhibit characteristics such as genomic instability, loss of protein homeostasis, metabolic dysregulation, and epigenetic alterations. Direct cross-talk between senescent T cells and other immune cells further exacerbates the immunosuppressive TME. This immune-tumor cell interaction within the TME contributes to impaired tumor antigen recognition and surveillance by T cells. The presence of senescent T cells is often associated with poor prognosis and reduced efficacy of immunotherapies; thus, targeting the tumor-promoting mechanisms of T-cell senescence may provide novel insights into improving tumor immunotherapy and patient outcomes. This review explores the contributors to tumor-derived T-cell senescence, the link between T-cell senescence and tumor prognosis, and the potential for targeting T-cell senescence to enhance tumor immunotherapy.
Collapse
Affiliation(s)
- Lihaoyun Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lingxuan Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weiming Mou
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Kai Miao
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
2
|
Khan M, Alteneder M, Reiter W, Krausgruber T, Dobnikar L, Madern M, Waldherr M, Bock C, Hartl M, Ellmeier W, Henriksson J, Boucheron N. Single-cell and chromatin accessibility profiling reveals regulatory programs of pathogenic Th2 cells in allergic asthma. Nat Commun 2025; 16:2565. [PMID: 40089475 PMCID: PMC11910648 DOI: 10.1038/s41467-025-57590-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Lung pathogenic T helper type 2 (pTh2) cells are important in mediating allergic asthma, but fundamental questions remain regarding their heterogeneity and epigenetic regulation. Here we investigate immune regulation in allergic asthma by single-cell RNA sequencing in mice challenged with house dust mite, in the presence and absence of histone deacetylase 1 (HDAC1) function. Our analyses indicate two distinct highly proinflammatory subsets of lung pTh2 cells and pinpoint thymic stromal lymphopoietin (TSLP) and Tumour Necrosis Factor Receptor Superfamily (TNFRSF) members as important drivers to generate pTh2 cells in vitro. Using our in vitro model, we uncover how signalling via TSLP and a TNFRSF member shapes chromatin accessibility at the type 2 cytokine gene loci by modulating HDAC1 repressive function. In summary, we have generated insights into pTh2 cell biology and establish an in vitro model for investigating pTh2 cells that proves useful for discovering molecular mechanisms involved in pTh2-mediated allergic asthma.
Collapse
Affiliation(s)
- Matarr Khan
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Marlis Alteneder
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Wolfgang Reiter
- Max Perutz Labs, Mass Spectrometry Facility, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Center for Medical Data Science, Institute of Artificial Intelligence, Vienna, Austria
| | - Lina Dobnikar
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Moritz Madern
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Monika Waldherr
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
- FH Campus Wien, Department of Applied Life Sciences/Bioengineering/Bioinformatics, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Center for Medical Data Science, Institute of Artificial Intelligence, Vienna, Austria
| | - Markus Hartl
- Max Perutz Labs, Mass Spectrometry Facility, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Vienna, Austria
| | - Wilfried Ellmeier
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Johan Henriksson
- Umeå University, Umeå Centre for Microbial Research (UCMR), Integrated Science Lab (Icelab), Department of Molecular Biology, Umeå, Sweden
| | - Nicole Boucheron
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria.
| |
Collapse
|
3
|
Zhao W, Yao Y, Li Q, Xue Y, Gao X, Liu X, Zhang Q, Zheng J, Sun S. Molecular mechanism of co-stimulatory domains in promoting CAR-T cell anti-tumor efficacy. Biochem Pharmacol 2024; 227:116439. [PMID: 39032532 DOI: 10.1016/j.bcp.2024.116439] [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] [Received: 05/14/2024] [Revised: 06/28/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Chimeric antigen receptor (CAR)-engineered T cells have been defined as 'living drug'. Adding a co-stimulatory domain (CSD) has enhanced the anti-hematological effects of CAR-T cells, thereby elevating their viability for medicinal applications. Various CSDs have helped prepare CAR-T cells to study anti-tumor efficacy. Previous studies have described and summarized the anti-tumor efficacy of CAR-T cells obtained from different CSDs. However, the underlying molecular mechanisms by which different CSDs affect CAR-T function have been rarely reported. The role of CSDs in T cells has been significantly studied, but whether they can play a unique role as a part of the CAR structure remains undetermined. Here, we summarized the effects of CSDs on CAR-T signaling pathways based on the limited references and speculated the possible mechanism depending on the specific characteristics of CAR-T cells. This review will help understand the molecular mechanism of CSDs in CAR-T cells that exert different anti-tumor effects while providing potential guidance for further interventions to enhance anti-tumor efficacy in immunotherapy.
Collapse
Affiliation(s)
- Wanxin Zhao
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qihong Li
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Xue
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoge Gao
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qing Zhang
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Shishuo Sun
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
4
|
He J, Zeng X, Wang C, Wang E, Li Y. Antibody-drug conjugates in cancer therapy: mechanisms and clinical studies. MedComm (Beijing) 2024; 5:e671. [PMID: 39070179 PMCID: PMC11283588 DOI: 10.1002/mco2.671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Antibody-drug conjugates (ADCs) consist of monoclonal antibodies that target tumor cells and cytotoxic drugs linked through linkers. By leveraging antibodies' targeting properties, ADCs deliver cytotoxic drugs into tumor cells via endocytosis after identifying the tumor antigen. This precise method aims to kill tumor cells selectively while minimizing harm to normal cells, offering safe and effective therapeutic benefits. Recent years have seen significant progress in antitumor treatment with ADC development, providing patients with new and potent treatment options. With over 300 ADCs explored for various tumor indications and some already approved for clinical use, challenges such as resistance due to factors like antigen expression, ADC processing, and payload have emerged. This review aims to outline the history of ADC development, their structure, mechanism of action, recent composition advancements, target selection, completed and ongoing clinical trials, resistance mechanisms, and intervention strategies. Additionally, it will delve into the potential of ADCs with novel markers, linkers, payloads, and innovative action mechanisms to enhance cancer treatment options. The evolution of ADCs has also led to the emergence of combination therapy as a new therapeutic approach to improve drug efficacy.
Collapse
Affiliation(s)
- Jun He
- Department of General Surgery Jiande Branch of the Second Affiliated Hospital, School of Medicine, Zhejiang University Jiande Zhejiang China
| | - Xianghua Zeng
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| | - Chunmei Wang
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| | - Enwen Wang
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| | - Yongsheng Li
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| |
Collapse
|
5
|
Kadin ME, Morgan J, Wei W, Song Z, Yang Y. CD30 Regulation of IL-13-STAT6 Pathway in Breast Implant-Associated Anaplastic Large Cell Lymphoma. Aesthet Surg J 2023; 43:137-146. [PMID: 35999655 PMCID: PMC10208747 DOI: 10.1093/asj/sjac234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) is a rare, usually indolent CD30+ T-cell lymphoma with tumor cells, often surrounded by eosinophils, expressing IL-13 and pSTAT6. OBJECTIVES The aim of this study was to understand the unique tumor pathology and growth regulation of BIA-ALCL, leading to potential targeted therapies. METHODS We silenced CD30 and analyzed its effect on IL-13 signaling and tumor cell viability. IL-13 signaling receptors of BIA-ALCL cell lines were evaluated by flow cytometry and pSTAT6 detected by immunohistochemistry. CD30 was deleted by CRISPR/Cas9 editing. Effects of CD30 deletion on transcription of IL-13 and IL-4, and phosphorylation of STAT6 were determined by real-time polymerase chain reaction and western blotting. The effect of CD30 deletion on p38 mitogen-activated protein kinase (MAPK) phosphorylation was determined. Suppression of IL-13 transcription by a p38 MAPK inhibitor was tested. Tumor cell viability following CD30 deletion and treatment with a pSTAT6 inhibitor were measured in cytotoxicity assays. RESULTS BIA-ALCL lines TLBR1 and TLBR2 displayed signaling receptors IL-4Rα, IL-13Rα1 and downstream pSTAT6. Deletion of CD30 by CRISPR/Cas9 editing significantly decreased transcription of IL-13, less so Th2 cytokine IL-4, and phosphorylation of STAT6. Mechanistically, we found CD30 expression is required for p38 MAPK phosphorylation and activation, and IL-13-STAT6 signaling was reduced by an inhibitor of p38 MAPK in BIA-ALCL tumor cells. Tumor cell viability was decreased by silencing of CD30, and a specific inhibitor of STAT6, indicating STAT6 inhibition is cytotoxic to BIA-ALCL tumor cells. CONCLUSIONS These findings suggest reagents targeting the IL-13 pathway, pSTAT6 and p38 MAPK, may become useful for treating BIA-ALCL patients.
Collapse
Affiliation(s)
- Marshall E Kadin
- Department of Pathology and Laboratory Medicine, Brown University Alpert
School of Medicine, Providence, RI. USA
| | | | - Wei Wei
- Fox Chase Cancer Center, Philadelphia,
PA, USA
| | - Zhihui Song
- Fox Chase Cancer Center, Philadelphia,
PA, USA
| | - Yibin Yang
- Blood Cell Development and Function Program, Fox Chase Cancer
Center, Philadelphia, PA, USA
| |
Collapse
|
6
|
Nikolaenko L, Nademanee A. Brentuximab vedotin and its use in the treatment of advanced Hodgkin's lymphoma. Future Oncol 2020; 16:2273-2282. [PMID: 32677451 DOI: 10.2217/fon-2020-0026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Brentuximab vedotin (BV), a CD30-directed antibody-drug conjugate, is US FDA approved for treatment of classic Hodgkin lymphoma (cHL) after progression or relapse of at least two prior lines of chemotherapy or autologous stem cell transplantation, as consolidation therapy after autologous stem cell transplantation for high-risk patients and as a front-line therapy for previously untreated, advanced-stage cHL in combination with chemotherapy. BV is a well-tolerated treatment in previously heavily pretreated relapsed/refractory cHL and in treatment-naive patients. BV use, in combination with other antineoplastic agents for cHL, is under investigation in multiple prospective clinical trials.
Collapse
Affiliation(s)
- Liana Nikolaenko
- Department of Hematology/Bone Marrow Transplant, City of Hope, Duarte, CA 91010, USA
| | - Auayporn Nademanee
- Department of Hematology/Bone Marrow Transplant, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
7
|
Kadin ME, Hamilton RG, Vonderheid EC. Evidence linking atopy and staphylococcal superantigens to the pathogenesis of lymphomatoid papulosis, a recurrent CD30+ cutaneous lymphoproliferative disorder. PLoS One 2020; 15:e0228751. [PMID: 32049976 PMCID: PMC7015403 DOI: 10.1371/journal.pone.0228751] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/22/2020] [Indexed: 01/08/2023] Open
Abstract
Background Primary cutaneous CD30+ lymphoproliferative disorders (CD30CLPD) are the second most common type of cutaneous T cell lymphoma (CTCL) and include lymphomatoid papulosis (LyP) and primary cutaneous anaplastic large cell lymphoma (pcALCL). Case reports and small patient series suggest an association of CD30CLPD with atopic disorders. However, the prevalence of atopy in patients with CD30CLPD in retrospective studies depends on patients’ recall which is not always reliable. More objective criteria of atopy include evidence of skin reactivity to allergens (positive prick test) and evidence of allergen-specific IgE in serum. This study was undertaken to test the hypothesis that atopy is prevalent in patients with CD30CLPD using serologic criteria of allergen-specific IgE antibodies to aeroallergens and Staphylococcal aureus enterotoxin superantigens (SSAgs). Methods We tested serum samples of CD30CLPD for common IgE-specific airborne allergens with the Phadiatop test, which if positive, is regarded as serologic evidence of atopy in adults. Sera were also tested for IgE antibodies reactive to three Staphylococcal enterotoxins with superantigenic properties (SSAg-IgE). Control sera were obtained from adult subjects evaluated for rhino-sinusitis and a negative Phadiatop test. Patients’ history of an atopic disorder was obtained by retrospective chart review. Findings Nearly 50% of patients with the most common LyP types (A and C) had a positive Phadiatop test for allergic sensitization to common airborne allergens, and total serum IgE (IgE-t) was increased compared to non-atopic controls. At the IgE antibody concentration generally used to define serologic atopy (≥ 0.35 kUA/L), 8/31 (26%) samples of CD30CLPD and 7/28 (25%) samples of LyP were reactive to at least one SSAg-IgE compared to 3/52 (6%) control specimens (P = 0.016 and P = 0.028, respectively). TSST1-IgE was detected in 7 (23%) specimens of CD30CLPD, often together with SEB-IgE; SEA-IgE ≥ 0.35 kUA/L was not detected. For control specimens, TSST1-IgE exceeded the 0.35 kUA/L threshold in 3 (6%) specimens. Conclusions Patients with LyP types A and C have serologic evidence of atopy against common airborne antigens and SSAgs when compared to control adult subjects who had rhino-sinusitis and a negative Phadiatop test for aero-IgEs. Serologic evidence of atopy exceeded that determined by LyP patients’ personal history. The findings support our hypothesis that an atopic diathesis may contribute to the pathogenesis of the most common types of LyP (A and C).
Collapse
Affiliation(s)
- Marshall E. Kadin
- Department of Dermatology, Boston University, Boston, MA, United States of America
- Department of Pathology and Laboratory Medicine, Lifespan-Rhode Island Hospital, Providence, RI, United States of America
- Department of Dermatology and Skin Surgery, Roger Williams Medical Center, Providence, RI, United States of America
- * E-mail:
| | - Robert G. Hamilton
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- Johns Hopkins Dermatology, Allergy and Clinical Immunology Reference Laboratory, Baltimore, MD, United States of America
| | - Eric C. Vonderheid
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- Sydney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
8
|
Cell of Origin and Immunologic Events in the Pathogenesis of Breast Implant-Associated Anaplastic Large-Cell Lymphoma. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 190:2-10. [PMID: 31610171 PMCID: PMC7298558 DOI: 10.1016/j.ajpath.2019.09.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/10/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022]
Abstract
Breast implant–associated anaplastic large-cell lymphoma (BIA-ALCL) is a CD30-positive, anaplastic lymphoma kinase–negative T-cell lymphoma. Nearly all cases have been associated with textured implants. Most cases are of effusion-limited, indolent disease, with an excellent prognosis after implant and capsule removal. However, capsular invasion and tumor mass have a more aggressive course and a fatal outcome risk. This review summarizes the current knowledge on BIA-ALCL cell of origin and immunologic factors underlying its pathogenesis. Cytokine expression profiling of BIA-ALCL cell lines and clinical specimens reveals a predominantly type 17 helper T-cell (Th17)/Th1 signature, implicating this as its cell of origin. However, a Th2 allergic inflammatory response is suggested by the presence of IL-13, with infiltration of eosinophils and IgE-coated mast cells in clinical specimens of BIA-ALCL. The microenvironment-induced T-cell plasticity, a factor increasingly appreciated, may partially explain these divergent results. Mutations resulting in constitutive Janus kinase (JAK)–STAT activation have been detected and associated with BIA-ALCL pathogenesis in a small number of cases. One possible scenario is that an inflammatory microenvironment stimulates an immune response, followed by polyclonal expansion of Th17/Th1 cell subsets with release of inflammatory cytokines and chemokines and accumulation of seroma. JAK-STAT3 gain-of-function mutations within this pathway and others may subsequently lead to monoclonal T-cell proliferation and clinical BIA-ALCL. Current research suggests that therapies targeting JAK proteins warrant investigation in BIA-ALCL.
Collapse
|
9
|
Bishop GA, Stunz LL, Hostager BS. TRAF3 as a Multifaceted Regulator of B Lymphocyte Survival and Activation. Front Immunol 2018; 9:2161. [PMID: 30319624 PMCID: PMC6165887 DOI: 10.3389/fimmu.2018.02161] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 09/03/2018] [Indexed: 12/20/2022] Open
Abstract
The adaptor protein TNF receptor-associated factor 3 (TRAF3) serves as a powerful negative regulator in multiple aspects of B cell biology. Early in vitro studies in transformed cell lines suggested the potential of TRAF3 to inhibit signaling by its first identified binding receptor, CD40. However, because the canonical TRAF3 binding site on many receptors also mediates binding of other TRAFs, and whole-mouse TRAF3 deficiency is neonatally lethal, an accurate understanding of TRAF3's specific functions was delayed until conditional TRAF3-deficient mice were produced. Studies of B cell-specific TRAF3-deficient mice, complemented by investigations in normal and malignant mouse and human B cells, reveal that TRAF3 has powerful regulatory roles that are unique to this TRAF, as well as functions context-specific to the B cell. This review summarizes the current state of knowledge of these roles and functions. These include inhibition of signaling by plasma membrane receptors, negative regulation of intracellular receptors, and restraint of cytoplasmic NF- κB pathways. TRAF3 is also now known to function as a resident nuclear protein, and to impact B cell metabolism. Through these and additional mechanisms TRAF3 exerts powerful restraint upon B cell survival and activation. It is thus perhaps not surprising that TRAF3 has been revealed as an important tumor suppressor in B cells. The many and varied functions of TRAF3 in B cells, and new directions to pursue in future studies, are summarized and discussed here.
Collapse
Affiliation(s)
- Gail A. Bishop
- Department of Microbiology & Immunology, University of Iowa, Iowa City, IA, United States
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
- Iowa City VA Health Care System, Iowa City, Iowa City, IA, United States
| | - Laura L. Stunz
- Department of Microbiology & Immunology, University of Iowa, Iowa City, IA, United States
| | - Bruce S. Hostager
- Department of Microbiology & Immunology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
10
|
Chen Z, Guan P, Shan T, Ye Y, Gao L, Wang Z, Zhao S, Zhang W, Zhang L, Pan L, Liu W. CD30 expression and survival in extranodal NK/T-cell lymphoma: a systematic review and meta-analysis. Oncotarget 2018; 9:16547-16556. [PMID: 29662666 PMCID: PMC5893261 DOI: 10.18632/oncotarget.24044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/02/2018] [Indexed: 02/05/2023] Open
Abstract
Background The paradoxical reports about the prognostic value of the CD30 expression in extranodal NK/T-cell lymphoma (ENKTL) have restricted its further applications in clinical practice. To identify the common effects and the variation, we conducted this systematic review and meta-analysis. Methods PubMed, MEDLINE, Embase, and Web of Science were searched between January 1975 and 31 January 2017. The pooled hazard ratio was used to estimate the effect of the CD30 expression on overall survival. Bias was assessed by prespecified criteria referring to Reporting Recommendations for Tumor Marker Prognostic Studies and Newcastle-Ottawa Scale. Results Ten retrospective cohort studies with 310 patients are included. CD30 is associated with better overall survival significantly (HR 0.71, 95% CI 0.51 to 0.99, I2 = 0%). A greater effect is observed among studies including participants predominant in regional involvement (HR 0.31, 95%CI 0.13 to 0.76, I2 = 0%) compared with those in systemic involvement. Conclusions This study indicates that the CD30 expression is significantly associated with better prognosis in ENKTL, especially for patients with regional lymphoma involvement.
Collapse
Affiliation(s)
- Zihang Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.,Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Pujun Guan
- Huaxi Magnetic Resonance Research Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.,Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tong Shan
- Institute of Public Health, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Yunxia Ye
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Limin Gao
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhi Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Sha Zhao
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenyan Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ling Pan
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weiping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
11
|
Fernández-Ruiz M, Parra P, López-Medrano F, Ruiz-Merlo T, González E, Polanco N, Origüen J, San Juan R, Andrés A, Aguado JM. Serum sCD30: A promising biomarker for predicting the risk of bacterial infection after kidney transplantation. Transpl Infect Dis 2017; 19. [PMID: 28122147 DOI: 10.1111/tid.12668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/08/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND The transmembrane glycoprotein CD30 contributes to regulate the balance between Th1 and Th2 responses. Previous studies have reported conflicting results on the utility of its soluble form (sCD30) to predict post-transplant infection. METHODS Serum sCD30 was measured by a commercial ELISA assay at baseline and post-transplant months 1, 3, and 6 in 100 kidney transplant (KT) recipients (279 monitoring points). The impact of sCD30 levels on the incidence of overall, bacterial and opportunistic infection during the first 12 months after transplantation was assessed by Cox regression. RESULTS There were no differences in serum sCD30 according to the occurrence of overall or opportunistic infection. However, sCD30 levels were higher in patients with bacterial infection compared to those without at baseline (P=.038) and months 1 (P<.0001), 3 (P=.043), and 6 after transplantation (P=.006). Patients with baseline sCD30 levels ≥13.5 ng/mL had lower 12-month bacterial infection-free survival (35.0% vs 80.0%; P<.0001). After adjusting for potential confounders, baseline sCD30 levels ≥13.5 ng/mL remained as an independent risk factor for bacterial infection (adjusted hazard ratio [aHR]: 4.65; 95% confidence interval [CI]: 2.05-10.53; <.001). Analogously, sCD30 levels ≥6.0 ng/mL at month 1 acted as a risk factor for subsequent bacterial infection (aHR: 5.29; 95% CI: 1.11-25.14; P=.036). CONCLUSION Higher serum sCD30 levels were associated with an increased risk of bacterial infection after KT. We hypothesize that this biomarker reflects a Th2 -polarized T-cell response, which exerts a detrimental effect on the immunity against bacterial pathogens.
Collapse
Affiliation(s)
- Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Patricia Parra
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Francisco López-Medrano
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Tamara Ruiz-Merlo
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Esther González
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Natalia Polanco
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Julia Origüen
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Rafael San Juan
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Amado Andrés
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| |
Collapse
|
12
|
Tsuyama N, Sakamoto K, Sakata S, Dobashi A, Takeuchi K. Anaplastic large cell lymphoma: pathology, genetics, and clinical aspects. J Clin Exp Hematop 2017; 57:120-142. [PMID: 29279550 PMCID: PMC6144189 DOI: 10.3960/jslrt.17023] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 11/19/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022] Open
Abstract
Anaplastic large cell lymphoma (ALCL) was first described in 1985 as a large-cell neoplasm with anaplastic morphology immunostained by the Ki-1 antibody, which recognizes CD30. In 1994, the nucleophosmin (NPM)-anaplastic lymphoma kinase (ALK) fusion receptor tyrosine kinase was identified in a subset of patients, leading to subdivision of this disease into ALK-positive and -negative ALCL in the present World Health Organization classification. Due to variations in morphology and immunophenotype, which may sometimes be atypical for lymphoma, many differential diagnoses should be considered, including solid cancers, lymphomas, and reactive processes. CD30 and ALK are key molecules involved in the pathogenesis, diagnosis, and treatment of ALCL. In addition, signal transducer and activator of transcription 3 (STAT3)-mediated mechanisms are relevant in both types of ALCL, and fusion/mutated receptor tyrosine kinases other than ALK have been reported in ALK-negative ALCL. ALK-positive ALCL has a better prognosis than ALK-negative ALCL or other peripheral T-cell lymphomas. Patients with ALK-positive ALCL are usually treated with anthracycline-based regimens, such as combination cyclophosphamide, doxorubicin, vincristine, and prednisolone (CHOP) or CHOEP (CHOP plus etoposide), which provide a favorable prognosis, except in patients with multiple International Prognostic Index factors. For targeted therapies, an anti-CD30 monoclonal antibody linked to a synthetic antimitotic agent (brentuximab vedotin) and ALK inhibitors (crizotinib, alectinib, and ceritinib) are being used in clinical settings.
Collapse
|
13
|
Amarapurkar P, Rosenblatt JD, Pereira D. Brentuximab: a major advance in treatment of CD30-positive malignancies. Int J Hematol Oncol 2015. [DOI: 10.2217/ijh.15.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Antibody-directed therapies allow greater selectivity in targeting of tumor associated antigens and spare normal cells. Brentuximab vedotin is an anti-CD30 antibody–drug conjugate. It has demonstrated impressive activity in the treatment of refractory and or relapsed Hodgkin's lymphoma, anaplastic large cell lymphoma and other CD30+ lymphoid malignancies. Several ongoing trials are testing the potential use of brentuximab vedotin for treatment of various CD30+ and CD30- malignancies in the setting of high-risk untreated disease. It is being tested in combination with chemotherapy, and testing in combination with immune therapy is also planned. CD30 plays a pivotal role in immune regulation and is also an attractive new target for intervention in the setting of select auto-immune diseases, as well as graft versus host disease.
Collapse
Affiliation(s)
- Pooja Amarapurkar
- Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Joseph D Rosenblatt
- Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, 1475 NW 12th Avenue Miami, FL 33136, USA
| | - Denise Pereira
- Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, 1475 NW 12th Avenue Miami, FL 33136, USA
| |
Collapse
|
14
|
Inghirami G, Chan WC, Pileri S. Peripheral T-cell and NK cell lymphoproliferative disorders: cell of origin, clinical and pathological implications. Immunol Rev 2015; 263:124-59. [PMID: 25510275 DOI: 10.1111/imr.12248] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T-cell lymphoproliferative disorders are a heterogeneous group of neoplasms with distinct clinical-biological properties. The normal cellular counterpart of these processes has been postulated based on functional and immunophenotypic analyses. However, T lymphocytes have been proven to be remarkably capable of modulating their properties, adapting their function in relationship with multiple stimuli and to the microenvironment. This impressive plasticity is determined by the equilibrium among a pool of transcription factors and by DNA chromatin regulators. It is now proven that the acquisition of specific genomic defects leads to the enforcement/activation of distinct pathways, which ultimately alter the preferential activation of defined regulators, forcing the neoplastic cells to acquire features and phenotypes distant from their original fate. Thus, dissecting the landscape of the genetic defects and their functional consequences in T-cell neoplasms is critical not only to pinpoint the origin of these tumors but also to define innovative mechanisms to re-adjust an unbalanced state to which the tumor cells have become addicted and make them vulnerable to therapies and targetable by the immune system. In our review, we briefly describe the pathological and clinical aspects of the T-cell lymphoma subtypes as well as NK-cell lymphomas and then focus on the current understanding of their pathogenesis and the implications on diagnosis and treatment.
Collapse
Affiliation(s)
- Giorgio Inghirami
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino, Italy; Department of Pathology, and NYU Cancer Center, New York University School of Medicine, New York, NY, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | | | | |
Collapse
|
15
|
So T, Nagashima H, Ishii N. TNF Receptor-Associated Factor (TRAF) Signaling Network in CD4 + T-Lymphocytes. TOHOKU J EXP MED 2015; 236:139-54. [DOI: 10.1620/tjem.236.139] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Takanori So
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine
| | - Hiroyuki Nagashima
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine
| |
Collapse
|
16
|
Abstract
The FDA recently approved an agonistic anti-CD30 drug conjugate, Brentuximab vedotin, for the treatment for CD30-positive lymphomas. The potent clinical activity of Brentuximab vedotin in Hodgkin's lymphoma and anaplastic large-cell lymphoma was greeted with great enthusiasm by oncologists as it provided a new treatment modality for these diseases. In this review, we will describe how we obtained the hybridoma by pursuing a basic research experiment unrelated to CD30. I will also review what we know about the normal biological functions of CD30 that were studied primarily in murine models of disease but also in patients. The picture emerging is that one of the primary functions of CD30 is the control of memory cells providing costimulation and trafficking information or inducing apoptosis in a microenvironment and cytokine milieu-dependent manner.
Collapse
|
17
|
Abstract
T cells are essential for immune defenses against pathogens, such that viability of naïve T cells before antigen encounter is critical to preserve a polyclonal repertoire and prevent immunodeficiencies. The viability of naïve T cells before antigen recognition is ensured by IL-7, which drives expression of the prosurvival factor Bcl-2. Quiescent naïve T cells have low basal activity of the transcription factor NF-κB, which was assumed to have no functional consequences. In contrast to this postulate, our data show that basal nuclear NF-κB activity plays an important role in the transcription of IL-7 receptor α-subunit (CD127), enabling responsiveness of naïve T cells to the prosurvival effects of IL-7 and allowing T-cell persistence in vivo. Moreover, we show that this property of basal NF-κB activity is shared by mouse and human naïve T cells. Thus, NF-κB drives a distinct transcriptional program in T cells before antigen encounter by controlling susceptibility to IL-7. Our results reveal an evolutionarily conserved role of NF-κB in T cells before antigenic stimulation and identify a novel molecular pathway that controls T-cell homeostasis.
Collapse
|
18
|
Abstract
Co-stimulatory and co-inhibitory receptors have a pivotal role in T cell biology, as they determine the functional outcome of T cell receptor (TCR) signalling. The classic definition of T cell co-stimulation continues to evolve through the identification of new co-stimulatory and co-inhibitory receptors, the biochemical characterization of their downstream signalling events and the delineation of their immunological functions. Notably, it has been recently appreciated that co-stimulatory and co-inhibitory receptors display great diversity in expression, structure and function, and that their functions are largely context dependent. Here, we focus on some of these emerging concepts and review the mechanisms through which T cell activation, differentiation and function is controlled by co-stimulatory and co-inhibitory receptors.
Collapse
Affiliation(s)
- Lieping Chen
- Department of Immunobiology and Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06519, USA.
| | | |
Collapse
|
19
|
Fearnley LG, Nielsen LK. PATHLOGIC-S: a scalable Boolean framework for modelling cellular signalling. PLoS One 2012; 7:e41977. [PMID: 22879903 PMCID: PMC3413702 DOI: 10.1371/journal.pone.0041977] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/27/2012] [Indexed: 11/18/2022] Open
Abstract
Curated databases of signal transduction have grown to describe several thousand reactions, and efficient use of these data requires the development of modelling tools to elucidate and explore system properties. We present PATHLOGIC-S, a Boolean specification for a signalling model, with its associated GPL-licensed implementation using integer programming techniques. The PATHLOGIC-S specification has been designed to function on current desktop workstations, and is capable of providing analyses on some of the largest currently available datasets through use of Boolean modelling techniques to generate predictions of stable and semi-stable network states from data in community file formats. PATHLOGIC-S also addresses major problems associated with the presence and modelling of inhibition in Boolean systems, and reduces logical incoherence due to common inhibitory mechanisms in signalling systems. We apply this approach to signal transduction networks including Reactome and two pathways from the Panther Pathways database, and present the results of computations on each along with a discussion of execution time. A software implementation of the framework and model is freely available under a GPL license.
Collapse
Affiliation(s)
- Liam G. Fearnley
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Lars K. Nielsen
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
20
|
Abstract
Brentuximab vedotin (SGN-35; Adcetris®) is an anti-CD30 antibody conjugated via a protease-cleavable linker to the potent anti-microtubule agent monomethyl auristatin E (MMAE). Following binding to CD30, brentuximab vedotin is rapidly internalized and transported to lysosomes where MMAE is released and binds to tubulin, leading to cell cycle arrest and apoptosis. Several trials have shown durable antitumor activity with a manageable safety profile in patients with relapsed/refractory Hodgkin lymphoma, systemic anaplastic large cell lymphoma, or primary cutaneous CD30-positive lymphoproliferative disorders. Peripheral sensory neuropathy is a significant adverse event associated with brentuximab vedotin administration. Neuropathy symptoms are cumulative and dose-related. Multiple ongoing trials are currently evaluating brentuximab vedotin alone or in combination with other agents in relapsed/refractory patients, as well as patients with newly diagnosed disease.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA USA.
| | | |
Collapse
|
21
|
Hildebrand JM, Yi Z, Buchta CM, Poovassery J, Stunz LL, Bishop GA. Roles of tumor necrosis factor receptor associated factor 3 (TRAF3) and TRAF5 in immune cell functions. Immunol Rev 2012; 244:55-74. [PMID: 22017431 DOI: 10.1111/j.1600-065x.2011.01055.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A large and diverse group of receptors utilizes the family of cytoplasmic signaling proteins known as tumor necrosis factor receptor (TNFR)-associated factors (TRAFs). In recent years, there has been a resurgence of interest and exploration of the roles played by TRAF3 and TRAF5 in cellular regulation, particularly in cells of the immune system, the cell types of focus in this review. This work has revealed that TRAF3 and TRAF5 can play diverse roles for different receptors even in the same cell type, as well as distinct roles in different cell types. Evidence indicates that TRAF3 and TRAF5 play important roles beyond the TNFR-superfamily (SF) and viral mimics of its members, mediating certain innate immune receptor and cytokine receptor signals, and most recently, signals delivered by the T-cell receptor (TCR) signaling complex. Additionally, much research has demonstrated the importance of TRAF3-mediated cellular regulation via its cytoplasmic interactions with additional signaling proteins. In particular, we discuss below evidence for the participation by TRAF3 in a number of the regulatory post-translational modifications involving ubiquitin that are important in various signaling pathways.
Collapse
Affiliation(s)
- Joanne M Hildebrand
- Department of Microbiology, The University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | |
Collapse
|
22
|
The role of costimulatory receptors of the tumour necrosis factor receptor family in atherosclerosis. J Biomed Biotechnol 2011; 2012:464532. [PMID: 22235167 PMCID: PMC3253462 DOI: 10.1155/2012/464532] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 10/11/2011] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that is mediated by both the innate and adaptive immune responses. T lymphocytes, that together with B cells are the cellular effectors of the adaptive immune system, are currently endowed with crucial roles in the development and progression of atherosclerosis. Costimulatory receptors are a class of molecules expressed by T lymphocytes that regulate the activation of T cells and the generation of effector T-cell responses. In this review we present the roles of costimulatory receptors of the tumour necrosis factor receptor (TNFR) superfamily in atherosclerosis and discuss the implications for future therapies that could be used to specifically modulate the immune response of pathogenic T cells in this disease.
Collapse
|
23
|
Fuchiwaki T, Sun X, Fujimura K, Yamada H, Shibata K, Muta H, Podack ER, Kawauchi H, Yoshikai Y. The central role of CD30L/CD30 interactions in allergic rhinitis pathogenesis in mice. Eur J Immunol 2011; 41:2947-54. [PMID: 21739429 DOI: 10.1002/eji.201141423] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Revised: 05/17/2011] [Accepted: 06/27/2011] [Indexed: 12/27/2022]
Abstract
CD30 ligand (CD30L) plays an important role in the amplification and/or activation of effector CD4(+) T cells, irrespective of Th cell subset. To examine the role of CD30L in allergic rhinitis, we evaluated an OVA model of allergic rhinitis in CD30L knock out (KO) mice on a BALB/c background sensitized with OVA. Symptoms of allergic rhinitis such as eosinophil infiltration into the nasal mucosa were drastically diminished in OVA-sensitized CD30L KO mice following intranasal challenge with OVA. The levels of OVA-specific IgE in the sera and the Th2 response in nasopharynx-associated lymphoid tissues and cervical LNs of CD30L KO mice were significantly lower than those of WT mice following intranasal challenge with OVA. Intranasal administration of CD30-Ig during the effector phase with OVA significantly prevented the development of allergic rhinitis in WT mice. These results suggest that CD30L plays an important role in allergic rhinitis and that the inhibition of CD30L/CD30 signaling might be useful as a novel biological therapy for allergic rhinitis.
Collapse
Affiliation(s)
- Takafumi Fuchiwaki
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Deutsch YE, Tadmor T, Podack ER, Rosenblatt JD. CD30: an important new target in hematologic malignancies. Leuk Lymphoma 2011; 52:1641-54. [DOI: 10.3109/10428194.2011.574761] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
25
|
Satpathy S, Shenoy GN, Kaw S, Vaidya T, Bal V, Rath S, George A. Inhibition of terminal differentiation of B cells mediated by CD27 and CD40 involves signaling through JNK. THE JOURNAL OF IMMUNOLOGY 2010; 185:6499-507. [PMID: 20974987 DOI: 10.4049/jimmunol.0903229] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
B cells responding to cognate Ag in vivo undergo clonal expansion that is followed by differentiation into Ab-secreting plasma cells or into quiescent restimulable memory. Both these events occur in the germinal center and require that cells exit from proliferation, but the signals that lead to one or the other of these mutually exclusive differentiation pathways have not been definitively characterized. Previous experiments have shown that signals transduced through the TNFRs CD27 and CD40 at the time of B cell stimulation in vitro or in vivo can influence this cell fate decision by inhibiting terminal differentiation and promoting memory. In this study, we show that the PIQED domain of the cytoplasmic tail of murine CD27 and the adapter molecule TNFR-associated factor 2 are involved in this effect. Using pharmacological inhibitors of signaling intermediates, we identify JNK as being necessary and sufficient for the observed inhibition of terminal differentiation. While JNK is involved downstream of CD40, inhibition of the MEK pathway can also partially restore plasma cell generation, indicating that both signaling intermediates may be involved. We also show that inhibition of induction of IFN regulatory factor 4 and B lymphocyte induced maturation protein 1 are downstream events common to both receptors.
Collapse
|
26
|
Forero-Torres A, Leonard JP, Younes A, Rosenblatt JD, Brice P, Bartlett NL, Bosly A, Pinter-Brown L, Kennedy D, Sievers EL, Gopal AK. A Phase II study of SGN-30 (anti-CD30 mAb) in Hodgkin lymphoma or systemic anaplastic large cell lymphoma. Br J Haematol 2009; 146:171-9. [DOI: 10.1111/j.1365-2141.2009.07740.x] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Ho DS, Rea AJ, Abraham LJ. Functional aspects of the CD30 gene in Hodgkin’s lymphoma and anaplastic large cell lymphoma. Oncol Rev 2009. [DOI: 10.1007/s12156-009-0012-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
28
|
Baum CL, Stone MS, Liu V. Atypical intravascular CD30+ T-cell proliferation following trauma in a healthy 17-year-old male: first reported case of a potential diagnostic pitfall and literature review. J Cutan Pathol 2009; 36:350-4. [DOI: 10.1111/j.1600-0560.2008.01033.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Tang C, Yamada H, Shibata K, Muta H, Wajjwalku W, Podack ER, Yoshikai Y. A novel role of CD30L/CD30 signaling by T-T cell interaction in Th1 response against mycobacterial infection. THE JOURNAL OF IMMUNOLOGY 2009; 181:6316-27. [PMID: 18941223 DOI: 10.4049/jimmunol.181.9.6316] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A CD30 ligand (CD30L, CD153) is a type II membrane-associated glycoprotein belonging to the TNF family. To illustrate the potential role of CD30L in CD4(+) Th1 cell responses, we investigated the fate of Ag-specific CD4(+) T cells in CD30L-deficient (CD30L(-/-)) mice after Mycobacterium bovis bacillus Calmette-Guérin (BCG) infection. The number of bacteria was significantly higher in organs of CD30L(-/-) mice than in wild-type (WT) mice 4 wk postinfection. The numbers of purified protein derivative- or Ag85B-specific-IFN-gamma-producing-CD4(+) T cells in spleen, lung, or peritoneal exudate cells were significantly fewer in CD30L(-/-) mice than in WT mice. During the infection, CD30L was expressed mainly by CD44(+)CD3(+)CD4(+) T cells but not by CD3(+)CD8(+) T cells, B cells, dendritic cells, or macrophages. Costimulation with agonistic anti-CD30 mAb or coculturing with CD30L-transfected P815 cells restored IFN-gamma production by CD4(+) T cells from BCG-infected CD30L(-/-) mice. Coculturing with CD30L(+/+)CD4(+) T cells from BCG-infected WT mice also restored the number of IFN-gamma(+)CD30L(-/-)CD4(+) T cells. When transferred into the CD30L(+/+) mice, Ag-specific donor CD30L(-/-) CD4(+) T cells capable of producing IFN-gamma were restored to the compared level seen in CD30L(+/+) CD4(+) T cells on day 10 after BCG infection. When naive CD30L(+/+) T cells were transferred into CD30L(-/-) mice, IFN-gamma-producing-CD4(+) Th1 cells of donor origin were normally generated following BCG infection, and IFN-gamma-producing-CD30L(-/-)CD4(+) Th1 cells of host origin were partly restored. These results suggest that CD30L/CD30 signaling executed by CD30(+) T-CD30L(+) T cell interaction partly play a critical role in augmentation of Th1 response capable of producing IFN-gamma against BCG infection.
Collapse
Affiliation(s)
- Ce Tang
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
30
|
Saini D, Ramachandran S, Nataraju A, Benshoff N, Liu W, Desai N, Chapman W, Mohanakumar T. Activated effector and memory T cells contribute to circulating sCD30: potential marker for islet allograft rejection. Am J Transplant 2008; 8:1798-808. [PMID: 18786226 DOI: 10.1111/j.1600-6143.2008.02329.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
T-cell activation up-regulates CD30 resulting in an increase in serum soluble CD30 (sCD30). CD4+ T cells, a major source for sCD30, play a significant role in the pathogenesis of rejection. In this study, sCD30 was measured pre- and posttransplant in mouse islet allograft models and human islet allograft recipients. sCD30 was measured by ELISA in diabetic C57BL/6, CD4Knockout (KO) and CD8KO islet allograft recipients. sCD30 increased significantly prior to rejection (1.8 +/- 1 days) in 80% of allograft recipients. Sensitization with donor splenocytes, or a second graft, further increased sCD30 (282.5 +/- 53.5 for the rejecting first graft vs. 374.6 +/- 129 for the rejecting second graft) prior to rejection suggesting memory CD4+ T cells contribute to sCD30. CD4KO failed to reject islet allograft and did not demonstrate sCD30 increase. CD8KO showed elevated (227 +/- 107) sCD30 (1 day) prior to rejection. High pretransplant sCD30 (>20 U/ml) correlated with poor outcome in human islet allograft recipients. Further, increase in sCD30 posttransplant preceded (3-4 months) loss of islet function. We conclude that sCD30 is released from activated CD4 T cells prior to islet allograft rejection and monitoring sCD30 can be a valuable adjunct in the follow-up of islet transplant recipients.
Collapse
Affiliation(s)
- D Saini
- Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Stern JNH, Keskin DB, Zhang H, Lv H, Kato Z, Strominger JL. Amino acid copolymer-specific IL-10-secreting regulatory T cells that ameliorate autoimmune diseases in mice. Proc Natl Acad Sci U S A 2008; 105:5172-6. [PMID: 18362339 PMCID: PMC2278190 DOI: 10.1073/pnas.0712131105] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Indexed: 01/14/2023] Open
Abstract
IL-10-secreting regulatory T cell lines specific to glatiramer acetate [poly(Y,E,A,K)n] or poly(Y,F,A,K)n have been established from the enlarged spleen and lymph nodes that result from copolymer treatment of SJL mice in which experimental autoimmune encephalomyelitis was induced by PLP139-151. These CD4+CD25+T cell lines secrete high levels of IL-10 and IL-13 but only small amounts of IL-4 and virtually no TGF-beta, IL-17, IL-6, IFN-gamma, or TNF-alpha. Their phenotypes are particularly characterized by the absence of Foxp3 and the presence of two TNFR family members, CD30 and GITR. The lines proliferated specifically to the immunizing copolymers but were autoantigen-nonspecific, in that the same T cell line could suppress autoimmunity induced by three different autoantigens in SJL mice, i.e., PLP139-151(EAE), MBP85-99 (EAE), and bovine peripheral nerve myelin (experimental autoimmune neuritis), indicating they function by bystander suppression.
Collapse
MESH Headings
- Adoptive Transfer
- Amino Acids/immunology
- Animals
- Bystander Effect
- Cattle
- Cell Line
- Cell Proliferation
- Cell Separation
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Forkhead Transcription Factors/metabolism
- Interleukin-10/metabolism
- Mice
- Peptides/immunology
- Phenotype
- Receptors, Antigen, T-Cell/immunology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Joel N. H. Stern
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Derin B. Keskin
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Hong Zhang
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - HuiJuan Lv
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Zenichiro Kato
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Jack L. Strominger
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| |
Collapse
|
32
|
Molinero LL, Zhou P, Wang Y, Harlin H, Kee B, Abraham C, Alegre ML. Epidermal Langerhans cells promote skin allograft rejection in mice with NF-kappa B-impaired T cells. Am J Transplant 2008; 8:21-31. [PMID: 18021281 PMCID: PMC2829624 DOI: 10.1111/j.1600-6143.2007.02038.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
T cells play a major role in the acute rejection of transplanted organs. Using mice transgenic for a T-cell-restricted NF-kappaB super-repressor (IkappaBalphaDeltaN-Tg mice), we have previously shown that T-cell-NF-kappaB is essential for the acute rejection of cardiac but not skin allografts. In this study, we investigated the mechanism by which skin grafts activate IkappaBalphaDeltaN-Tg T cells. Rejection was not due to residual T-cell-NF-kappaB activity as mice with p50/p52(-/-) T cells successfully rejected skin grafts. Rather, skin but not cardiac allografts effectively induced proliferation of graft-specific IkappaBalphaDeltaN-Tg T cells. Rejection of skin grafts by IkappaBalphaDeltaN-Tg mice was in part dependent on the presence of donor Langerhans cells (LC), a type of epidermal dendritic cells (DC), as lack of LC in donor skin grafts resulted in prolongation of skin allograft survival and injection of LC at the time of cardiac transplantation was sufficient to promote cardiac allograft rejection by IkappaBalphaDeltaN-Tg mice. Our results suggest that LC allow NF-kappaB-impaired T cells to reach an activation threshold sufficient for transplant rejection. The combined blockade of T-cell-NF-kappaB with that of alternative pathways allowing activation of NF-kappaB-impaired T cells may be an effective strategy for tolerance induction to highly immunogenic organs.
Collapse
Affiliation(s)
- LL Molinero
- Department of Medicine, and Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - P Zhou
- Department of Medicine, and Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - Y Wang
- Department of Medicine, and Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - H Harlin
- Medical College of Wisconsin, WI 53226
| | - B Kee
- Department of Pathology, and Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - C Abraham
- Department of Medicine, and Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - ML Alegre
- Department of Medicine, and Committee on Immunology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
33
|
Nam SY, Kim YH, Do JS, Choi YH, Seo HJ, Yi HK, Hwang PH, Song CH, Lee HK, Kim JS, Podack ER. CD30 supports lung inflammation. Int Immunol 2007; 20:177-84. [PMID: 18089617 DOI: 10.1093/intimm/dxm130] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The physiological functions of CD30 have not been fully elucidated. Here we show that in CD30-deficient mice (CD30(-/-)), lung inflammation is significantly diminished in the ovalbumin (OVA) model of airway hyperreactivity. In CD30(-/-) mice, the recruitment of eosinophils into the airways after OVA-aerosol challenge of OVA-primed mice was significantly diminished when compared with wild-type (w.t.) mice. IL-13 levels were also significantly reduced in CD30(-/-) mice while levels of IFN-gamma, IL-4, IL-5 and IgE in bronchoalveolar lavage fluid, lung tissue and serum were comparable to w.t. mice. Peribronchial lymph node cells from CD30(-/-) mice, re-stimulated in vitro with OVA, secreted significantly lower levels of IL-13 than those from w.t. mice, but showed normal proliferative response and other cytokine production. Exogenous IL-13 reconstituted airway recruitment of leukocytes in OVA-challenged CD3O(-/-) mice. Adoptive transfer to naive w.t. mice of in vitro OVA-re-stimulated spleen cells from CD30(-/-) mice failed to induce eosinophilic pulmonary inflammation in contrast to transfer of primed cells from w.t. mice. These results indicate that CD30 is a regulator of T(h)2 responses in the effector-memory phase and a regulator of IL-13 production in memory cells in the lung.
Collapse
Affiliation(s)
- Sang-Yun Nam
- Department of Biological Science, School of Science and Technology, Jeonju University, Jeonju 560-759, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Saez-Rodriguez J, Simeoni L, Lindquist JA, Hemenway R, Bommhardt U, Arndt B, Haus UU, Weismantel R, Gilles ED, Klamt S, Schraven B. A logical model provides insights into T cell receptor signaling. PLoS Comput Biol 2007; 3:e163. [PMID: 17722974 PMCID: PMC1950951 DOI: 10.1371/journal.pcbi.0030163] [Citation(s) in RCA: 202] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Accepted: 07/05/2007] [Indexed: 12/15/2022] Open
Abstract
Cellular decisions are determined by complex molecular interaction networks. Large-scale signaling networks are currently being reconstructed, but the kinetic parameters and quantitative data that would allow for dynamic modeling are still scarce. Therefore, computational studies based upon the structure of these networks are of great interest. Here, a methodology relying on a logical formalism is applied to the functional analysis of the complex signaling network governing the activation of T cells via the T cell receptor, the CD4/CD8 co-receptors, and the accessory signaling receptor CD28. Our large-scale Boolean model, which comprises 94 nodes and 123 interactions and is based upon well-established qualitative knowledge from primary T cells, reveals important structural features (e.g., feedback loops and network-wide dependencies) and recapitulates the global behavior of this network for an array of published data on T cell activation in wild-type and knock-out conditions. More importantly, the model predicted unexpected signaling events after antibody-mediated perturbation of CD28 and after genetic knockout of the kinase Fyn that were subsequently experimentally validated. Finally, we show that the logical model reveals key elements and potential failure modes in network functioning and provides candidates for missing links. In summary, our large-scale logical model for T cell activation proved to be a promising in silico tool, and it inspires immunologists to ask new questions. We think that it holds valuable potential in foreseeing the effects of drugs and network modifications.
Collapse
Affiliation(s)
- Julio Saez-Rodriguez
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Luca Simeoni
- Institute of Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Rebecca Hemenway
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Ursula Bommhardt
- Institute of Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Boerge Arndt
- Institute of Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Utz-Uwe Haus
- Institute for Mathematical Optimization, Otto-von-Guericke University, Magdeburg, Germany
| | - Robert Weismantel
- Institute for Mathematical Optimization, Otto-von-Guericke University, Magdeburg, Germany
| | - Ernst D Gilles
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Steffen Klamt
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- * To whom correspondence should be addressed. E-mail: inquiries regarding the mathematical methodology should be addressed to Steffen Klamt, , and regarding the biological and experimental data to Burkhart Schraven,
| | - Burkhart Schraven
- Institute of Immunology, Otto-von-Guericke University, Magdeburg, Germany
- * To whom correspondence should be addressed. E-mail: inquiries regarding the mathematical methodology should be addressed to Steffen Klamt, , and regarding the biological and experimental data to Burkhart Schraven,
| |
Collapse
|
35
|
Abstract
Polyclonal antithymocyte globulins (AThG) are a subset of antilymphocyte antibody preparations derived from the sera of rabbits or horses immunized with unfractionated cells isolated from pediatric human thymi. In vivo, AThG preparations have been used to successfully treat antibody mediated rejection in kidney transplant recipients. In vitro, AThG can induce apoptosis of naïve and memory B cells and terminally differentiated plasma cells. The presence of B-cell reactive antibodies in AThG results from a thymic inoculum containing a significant percentage of CD20(+) B cells and CD138(+) plasma cells. In this paper, the experimental and clinical evidence supporting the B-cell activity of AThG preparations, and their mechanisms of action, are reviewed.
Collapse
Affiliation(s)
- Martin S Zand
- Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
36
|
Krysov SV, Rowley TF, Al-Shamkhani A. Inhibition of p38 mitogen-activated protein kinase unmasks a CD30-triggered apoptotic pathway in anaplastic large cell lymphoma cells. Mol Cancer Ther 2007; 6:703-11. [PMID: 17308066 DOI: 10.1158/1535-7163.mct-06-0544] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD30, a non-death domain-containing member of the tumor necrosis factor receptor superfamily, triggers apoptosis in anaplastic large cell lymphoma cells. The CD30 signaling pathways that lead to the induction of apoptosis are poorly defined. Here, we show that the induction of apoptosis by CD30 requires concurrent inhibition of p38 mitogen-activated protein kinase, which itself is activated by engagement of CD30 with CD30 ligand. Treatment of anaplastic large cell lymphoma cells with CD30 ligand and pharmacologic inhibitors of p38 mitogen-activated protein kinase, but not with CD30 ligand or inhibitors alone, triggered the activation of caspase-8 and the induction of apoptosis. Caspase-8 activation occurred within a few hours (2.5-4 h) after receptor triggering, was unaffected by the neutralization of ligands for the death domain-containing receptors TNFR1, Fas, DR3, DR4, or DR5, but was abolished by the expression of a dominant-negative form of the adaptor protein FADD. Importantly, we show that expression of the caspase-8 inhibitor c-FLIP(S) is strongly induced by the CD30 ligand, and that this is dependent on the activation of p38 mitogen-activated protein kinase. Thus, we provide evidence that the induction of apoptosis by CD30 in anaplastic large cell lymphoma cells is normally circumvented by the activation of p38 mitogen-activated protein kinase. These findings have implications for CD30-targeted immunotherapy of anaplastic large cell lymphoma.
Collapse
MESH Headings
- Apoptosis/drug effects
- Blotting, Western
- CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism
- Caspase 3/metabolism
- Caspase 8/metabolism
- Caspase Inhibitors
- Cell Cycle/drug effects
- Fas Ligand Protein/metabolism
- Fas-Associated Death Domain Protein/metabolism
- Humans
- Ki-1 Antigen/immunology
- Ki-1 Antigen/pharmacology
- Lymphoma, Large-Cell, Anaplastic/genetics
- Lymphoma, Large-Cell, Anaplastic/metabolism
- Lymphoma, Large-Cell, Anaplastic/pathology
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Member 25/metabolism
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Transfection
- Tumor Cells, Cultured/drug effects
- Tumor Necrosis Factor-alpha/metabolism
- fas Receptor/metabolism
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- p38 Mitogen-Activated Protein Kinases/genetics
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Sergey V Krysov
- Tenovus Research Laboratory, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, United Kingdom
| | | | | |
Collapse
|
37
|
Gao N, Schwartzberg P, Wilder JA, Blazar BR, Yuan D. B cell induction of IL-13 expression in NK cells: role of CD244 and SLAM-associated protein. THE JOURNAL OF IMMUNOLOGY 2006; 176:2758-64. [PMID: 16493031 DOI: 10.4049/jimmunol.176.5.2758] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
NK cells are an important component of the innate immune system that can also interact with B cells in a mutually productive manner. We have previously shown that activated B cells can induce NK cells to up-regulate their secretion of IFN-gamma. In this study, we show that B cells, and, particularly, marginal zone B cells, can, in addition, induce NK cells via direct cell-cell interactions to express mRNA encoding the Th2 cytokine IL-13. The induction of NK cell IL-13 mRNA expression requires the ligation of the CD244 receptor by the CD48 ligand on B cells via signaling pathways that depend upon expression of the X-linked lymphoproliferative disease gene product, SH2D1A/DSHP/SAP (SLAM-associated protein, or SAP) in NK cells. Thus, the positive signals attributed to the B cell activation of CD244 on murine NK cells appears to be more similar to the activity of CD244 on human cells. The induction of IL-13 mRNA by B cells may account for the effect of NK cells on the generation of Th2-type responses in the presence of some adjuvants.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/physiology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cell Communication/genetics
- Cell Communication/immunology
- Cells, Cultured
- Coculture Techniques
- Humans
- Interferon-gamma/biosynthesis
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interleukin-13/biosynthesis
- Interleukin-13/genetics
- Interleukin-13/physiology
- Intracellular Signaling Peptides and Proteins/deficiency
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/physiology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- RNA, Messenger/biosynthesis
- Receptors, Cell Surface/physiology
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/physiology
- Signaling Lymphocytic Activation Molecule Associated Protein
- Signaling Lymphocytic Activation Molecule Family
Collapse
Affiliation(s)
- Ning Gao
- Department of Molecular Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | |
Collapse
|
38
|
Dodeller F, Schulze-Koops H. The p38 mitogen-activated protein kinase signaling cascade in CD4 T cells. Arthritis Res Ther 2006; 8:205. [PMID: 16542479 PMCID: PMC1526596 DOI: 10.1186/ar1905] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Since the identification of the p38 mitogen-activated protein kinase (MAPK) as a key signal-transducing molecule in the expression of the proinflammatory cytokine tumor necrosis factor (TNF) more than 10 years ago, huge efforts have been made to develop inhibitors of p38 MAPK with the intent to modulate unwanted TNF activity in diseases such as autoimmune diseases or sepsis. However, despite some anti-inflammatory effects in animal models, no p38 MAPK inhibitor has yet demonstrated clinical efficacy in human autoimmune disorders. One possible reason for this paradox might relate to the fact that the p38 MAPK signaling cascade is involved in the functional regulation of several different cell types that all contribute to the complex pathogenesis of human autoimmune diseases. In particular, p38 MAPK has a multifaceted role in CD4 T cells that have been implicated in initiating and driving sustained inflammation in autoimmune diseases, such as rheumatoid arthritis or systemic vasculitis. Here we review recent advances in the understanding of the role of the p38 MAPK signaling cascade in CD4 T cells and the consequences that its inhibition provokes in T cell functions in vitro and in vivo. These new data suggest that p38 MAPK inhibitors may elicit several unwanted effects in human autoimmune diseases but may be useful for the treatment of allergic disorders.
Collapse
Affiliation(s)
- Francis Dodeller
- Nikolaus Fiebiger Center for Molecular Medicine, Clinical Research Group III, and Department of Internal Medicine III, University of Erlangen-Nuremberg, Glueckstrasse 6, 91054 Erlangen, Germany
| | - Hendrik Schulze-Koops
- Nikolaus Fiebiger Center for Molecular Medicine, Clinical Research Group III, and Department of Internal Medicine III, University of Erlangen-Nuremberg, Glueckstrasse 6, 91054 Erlangen, Germany
| |
Collapse
|
39
|
Watanabe M, Sasaki M, Itoh K, Higashihara M, Umezawa K, Kadin ME, Abraham LJ, Watanabe T, Horie R. JunB Induced by Constitutive CD30–Extracellular Signal-Regulated Kinase 1/2 Mitogen-Activated Protein Kinase Signaling Activates the CD30 Promoter in Anaplastic Large Cell Lymphoma and Reed-Sternberg Cells of Hodgkin Lymphoma. Cancer Res 2005; 65:7628-34. [PMID: 16140928 DOI: 10.1158/0008-5472.can-05-0925] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
High expression of CD30 and JunB is characteristic of tumor cells in anaplastic large cell lymphoma (ALCL) and Hodgkin lymphoma (HL). Possible interactions of CD30 and JunB were examined in this study. We found that the CD30 promoter in tumor cells of both nucleophosmin (NPM)-anaplastic lymphoma kinase (ALK)-positive and NPM-ALK-negative ALCL and HL is regulated by a constitutively active CD30-extracellular signal-regulated kinase (ERK) 1/2 mitogen-activated protein kinase (MAPK). Phosphorylation of ERK1/2 MAPK was confirmed in nuclei of tumor cells in both ALCL and HL. CD30-ERK1/2 MAPK signals induce JunB expression, which maintains high activity of the CD30 promoter. JunB induction seems to be largely independent of nuclear factor kappaB in ALCL and HL. These results show a common mechanism of CD30 overexpression in ALCL and HL, although the outcome of CD30 signaling differs between NPM-ALK-positive ALCL and NPM-ALK-negative ALCL, cutaneous ALCL, and HL as we recently reported.
Collapse
Affiliation(s)
- Mariko Watanabe
- Fourth Department of Internal Medicine, School of Medicine, Kitasato University, Sagamihara, Kanagawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Several members of the tumor necrosis factor receptor (TNFR) family function after initial T cell activation to sustain T cell responses. This review focuses on CD27, 4-1BB (CD137), OX40 (CD134), HVEM, CD30, and GITR, all of which can have costimulatory effects on T cells. The effects of these costimulatory TNFR family members can often be functionally, temporally, or spatially segregated from those of CD28 and from each other. The sequential and transient regulation of T cell activation/survival signals by different costimulators may function to allow longevity of the response while maintaining tight control of T cell survival. Depending on the disease condition, stimulation via costimulatory TNF family members can exacerbate or ameliorate disease. Despite these complexities, stimulation or blockade of TNFR family costimulators shows promise for several therapeutic applications, including cancer, infectious disease, transplantation, and autoimmunity.
Collapse
Affiliation(s)
- Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
41
|
Higuchi M, Matsuda T, Mori N, Yamada Y, Horie R, Watanabe T, Takahashi M, Oie M, Fujii M. Elevated expression of CD30 in adult T-cell leukemia cell lines: possible role in constitutive NF-kappaB activation. Retrovirology 2005; 2:29. [PMID: 15876358 PMCID: PMC1274245 DOI: 10.1186/1742-4690-2-29] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Accepted: 05/06/2005] [Indexed: 12/17/2022] Open
Abstract
Background Human T-cell leukemia virus type 1 (HTLV-1) is associated with the development of adult T-cell leukemia (ATL). HTLV-1 encoded Tax1 oncoprotein activates the transcription of genes involved in cell growth and anti-apoptosis through the NF-κB pathway, and is thought to play a critical role in the pathogenesis of ATL. While Tax1 expression is usually lost or minimal in ATL cells, these cells still show high constitutive NF-κB activity, indicating that genetic or epigenetic changes in ATL cells induce activation independent of Tax1. The aim of this study was to identify the molecules responsible for the constitutive activation of NF-κB in ATL cells using a retroviral functional cloning strategy. Results Using enhanced green fluorescent protein (EGFP) expression and blasticidin-resistance as selection markers, several retroviral cDNA clones exhibiting constitutive NF-κB activity in Rat-1 cells, including full-length CD30, were obtained from an ATL cell line. Exogenous stable expression of CD30 in Rat-1 cells constitutively activated NF-κB. Elevated expression of CD30 was identified in all ATL lines examined, and primary ATL cells from a small number of patients (8 out of 66 cases). Conclusion Elevated CD30 expression is considered one of the causes of constitutive NF-κB activation in ATL cells, and may be involved in ATL development.
Collapse
Affiliation(s)
- Masaya Higuchi
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Takehiro Matsuda
- Division of Molecular Virology and Oncology, Faculty of Medicine, University of the Ryukyus, Nishihara, Okinawa 903-0215, Japan
| | - Naoki Mori
- Division of Molecular Virology and Oncology, Faculty of Medicine, University of the Ryukyus, Nishihara, Okinawa 903-0215, Japan
| | - Yasuaki Yamada
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 825-8501, Japan
| | - Ryouichi Horie
- Fourth Department of Internal Medicine, Faculty of Medicine, Kitasato University, Sagamihara, Kanagawa 228-8555, Japan
| | - Toshiki Watanabe
- Laboratory of Tumor Cell Biology, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Minato-ku, Tokyo 108-109, Japan
| | - Masahiko Takahashi
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Masayasu Oie
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Masahiro Fujii
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| |
Collapse
|
42
|
Gilbert KM, Whitlow AB, Pumford NR. Environmental contaminant and disinfection by-product trichloroacetaldehyde stimulates T cells in vitro. Int Immunopharmacol 2004; 4:25-36. [PMID: 14975357 DOI: 10.1016/j.intimp.2003.10.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2003] [Revised: 08/14/2003] [Accepted: 10/03/2003] [Indexed: 01/28/2023]
Abstract
It had been shown previously that MRL+/+ mice exposed to occupationally relevant doses of the environmental contaminant trichloroethylene in their drinking water developed lupus-like symptoms and autoimmune hepatitis in association with activation of Interferon-gamma (IFN-gamma)-producing CD4+ T cells. Since trichloroethylene must be metabolized in order to promote the T-cell activation associated with autoimmunity, the present study was initiated to determine whether the immunoregulatory effects of trichloroethylene could be mimicked by one of its major metabolites, trichloroacetaldehyde (TCAA). At concentrations ranging from 0.04 to 1 mM TCAA co-stimulated proliferation of murine T-helper type 1 (Th1) cells treated with anti-CD3 antibody or antigen in vitro. TCAA at similar concentrations also induced phenotypic alterations commensurate with activation (upregulation of CD28 and downregulation of CD62L) in both cloned memory Th1 cells, as well as naïve CD4+ T cells from MRL+/+ mice. TCAA-induced Th1 cell activation was accompanied by phoshorylation of activating transcription factor 2 (ATF-2) and c-Jun, two components of the activator protein-1 (AP-1) transcription factor. TCAA at higher concentrations was also shown to form a Schiff base on T cells, and inhibition of Schiff base formation suppressed the ability of TCAA to phosphorylate ATF-2. Taken together, these results suggest that TCAA promotes T-cell activation via stimulation of the mitogen-activated protein (MAP) kinase pathway in association with Schiff base formation on T-cell surface proteins. By demonstrating that TCAA can stimulate T-cell function directly, these results may explain how the environmental toxicant trichloroethylene promotes T-cell activation and related autoimmunity in vivo.
Collapse
Affiliation(s)
- Kathleen M Gilbert
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences/Arkansas Children's Hospital Research Institute, 1120 Marshall Street, Little Rock, AR 72205, USA.
| | | | | |
Collapse
|
43
|
Abstract
Members of both the CD28 and TNFR families can have costimulatory roles in T cell activation. Gene targeted mice as well as in vivo blocking experiments have established distinct roles for CD28/B7; ICOS/ICOSL; CD27/CD70; 4-1BB/4-1BBL and OX40/OX40L during viral infection. Many issues remain to be addressed, including the timing and location of the interactions, the possibility of partial redundancy between related family members and the molecular basis for the specific phenotypes observed in the different gene targeted mice.
Collapse
Affiliation(s)
- Edward M Bertram
- Australian Phenomics Facility and Division of Immunology and Genetics, John Curtin School of Medical Research, Australian National University, Canberra, Australia 2601
| | | | | |
Collapse
|
44
|
Blazar BR, Levy RB, Mak TW, Panoskaltsis-Mortari A, Muta H, Jones M, Roskos M, Serody JS, Yagita H, Podack ER, Taylor PA. CD30/CD30 Ligand (CD153) Interaction Regulates CD4+T Cell-Mediated Graft-versus-Host Disease. THE JOURNAL OF IMMUNOLOGY 2004; 173:2933-41. [PMID: 15322151 DOI: 10.4049/jimmunol.173.5.2933] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD30, a TNFR family member, is expressed on activated CD4(+) and CD8(+) T cells and B cells and is a marker of Hodgkin's lymphoma; its ligand, CD30L (CD153) is expressed by activated CD4(+) and CD8(+) T cells, B cells, and macrophages. Signaling via CD30 can lead to proliferation or cell death. CD30-deficient (-/-) mice have impaired thymic negative selection and increased autoreactivity. Although human alloreactive T cells preferentially reside within the CD30(+) T cell subset, implicating CD30 as a regulator of T cell immune responses, the role of CD30/CD153 in regulating graft-vs-host disease (GVHD) has not been reported. We used a neutralizing anti-CD153 mAb, CD30(-/-) donor mice, and generated CD153(-/-) recipient mice to analyze the effect of CD30/CD153 interaction on GVHD induction. Our data indicate that the CD30/CD153 pathway is a potent regulator of CD4(+), but not CD8(+), T cell-mediated GVHD. Although blocking CD30/CD153 interactions in vivo did not affect alloreactive CD4(+) T cell proliferation or apoptosis, a substantial reduction in donor CD4(+) T cell migration into the gastrointestinal tract was readily observed with lesser effects in other GVHD target organs. Blockade of the CD30/CD153 pathway represents a new approach for preventing CD4(+) T cell-mediated GVHD.
Collapse
Affiliation(s)
- Bruce R Blazar
- Cancer Center and Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Leiro J, Arranz JA, Yáñez M, Ubeira FM, Sanmartín ML, Orallo F. Expression profiles of genes involved in the mouse nuclear factor-kappa B signal transduction pathway are modulated by mangiferin. Int Immunopharmacol 2004; 4:763-78. [PMID: 15135318 DOI: 10.1016/j.intimp.2004.03.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2003] [Revised: 03/01/2004] [Accepted: 03/02/2004] [Indexed: 01/03/2023]
Abstract
The polyphenol mangiferin (MA) has been shown to have various effects on macrophage function, including inhibition of phagocytic activity and of free radical production. To further characterize the immunomodulatory activity of MA, this study investigated its effects on expression by activated mouse macrophages of diverse genes related to the NF-kappaB signaling pathway, using a DNA hybridization array containing 96 NF-kappaB-related genes and on cytokine levels using a cytokine protein array. MA at 10 microM significantly inhibited the expression of (a) two genes of the Rel/NF-kappaB/IkappaB family, RelA and RelB (=I-rel), indicating an inhibitory effect on NF-kappaB-mediated signal transduction; (b) TNF receptor-associated factor 6 (Traf6), indicating probable blockage of activation of the NF-kappaB pathway by lipopolysaccharide (LPS), tumor necrosis factor (TNF), and interleukin 1 (IL-1); (c) other proteins involved in responses to TNF and in apoptotic pathways triggered by DNA damage, including the TNF receptor (TNF-R), the TNF-receptor-associated death domain (TRADD), and the receptor interacting protein (RIP); (d) the extracellular ligand IL-1alpha, again indicating likely interference with responses to IL-1; (e) the pro-inflammatory cytokines IL-1, IL-6, IL-12, TNF-alpha and RANTES (CCL5), and cytokines produced by monocytes and macrophages, including granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF); (f) other toll-like receptor proteins (in addition to Traf6), including JNK1, JNK2 and Tab1; (g) Scya2 (small inducible cytokine A2=monocyte chemoattractant protein 1); and (h) various intracellular adhesion molecules (ICAMs), and the vascular cell adhesion molecule VCAM-1, which is locally increased in atheromas. The inhibition of JNK1, together with stimulation of c-JUN (i.e. the Jun oncogene) and the previously reported superoxide-scavenging activity of MA, suggests that MA may protect cells against oxidative damage and mutagenesis. Taken together, these results indicate that MA modulates the expression of a large number of genes that are critical for the regulation of apoptosis, viral replication, tumorogenesis, inflammation and various autoimmune diseases, and raise the possibility that it may be of value in the treatment of inflammatory diseases and/or cancer.
Collapse
Affiliation(s)
- José Leiro
- Laboratorio de Parasitología, Instituto de Investigación y Análisis Alimentarios, Universidad de Santiago de Compostela, C/ Constantino Candeira s/n, 15782 Santiago de Compostela, La Coruña, Spain.
| | | | | | | | | | | |
Collapse
|
46
|
|
47
|
So T, Salek-Ardakani S, Nakano H, Ware CF, Croft M. TNF Receptor-Associated Factor 5 Limits the Induction of Th2 Immune Responses. THE JOURNAL OF IMMUNOLOGY 2004; 172:4292-7. [PMID: 15034043 DOI: 10.4049/jimmunol.172.7.4292] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The TNF receptor-associated factor (TRAF) family of molecules acts as adapter proteins for signaling pathways initiated by several members of the TNF receptor (TNFR) superfamily. TRAF5(-/-) animals are viable and have normal development of the immune system despite interacting with several TNFR family members. A clear role for TRAF5 has yet to emerge. OX40 (CD134) interacts with TRAF5, suggesting that this pathway could be involved in regulating T cell differentiation into Th1 or Th2 cells. In tissue culture, OX40 stimulation of TRAF5(-/-) T cells resulted in a pronounced Th2 phenotype with elevated levels of IL-4 and IL-5. Similarly, in vivo immunization with protein in adjuvant in the presence of an agonist anti-OX40 Ab resulted in enhanced Th2 development in TRAF5(-/-) mice. Additionally, lung inflammation induced by T cells, which is critically controlled by OX40, was more pronounced in TRAF5(-/-) mice, characterized by higher levels of Th2 cytokines. These results suggest that TRAF5 can limit the induction of Th2 responses, and that TRAF5 can play a role in modulating responses driven by OX40 costimulation.
Collapse
Affiliation(s)
- Takanori So
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, San Diego, CA 92121, USA
| | | | | | | | | |
Collapse
|
48
|
Dozmorov I, Saban MR, Knowlton N, Centola M, Saban R. Connective molecular pathways of experimental bladder inflammation. Physiol Genomics 2003; 15:209-22. [PMID: 12966137 DOI: 10.1152/physiolgenomics.00130.2003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inflammation is an inherent response of the organism that permits its survival despite constant environmental challenges. The process normally leads to recovery from injury and to healing. However, if targeted destruction and assisted repair are not properly phased, chronic inflammation can result in persistent tissue damage. To better understand the inflammatory process, we recently introduced a profiling methodology to identify common genes involved in bladder inflammation. The method represents a complementation to the classic quantification of inflammation and provides information regarding the early, intermediate, and late events in gene regulation. However, gene profiling fails to describe the molecular pathways and their interconnections involved in the particular inflammatory response. The present work introduces a new statistical technique for inferring functional interconnections between inflammatory pathways underlying classic models of bladder inflammation and permits the modeling of the inflammatory network. This new statistical method is based on variants of cluster analysis, Boolean networking, differential equations, Bayesian networking, and partial correlation. By applying partial correlation analysis, we developed mosaics of gene expression that permitted a global visualization of common and unique pathways elicited by different stimuli. The significance of these processes was tested from both biological and statistical viewpoints. We propose that connective mosaic may represent the necessary simplification step to visualize cDNA array results.
Collapse
Affiliation(s)
- Igor Dozmorov
- Oklahoma Medical Research Foundation, Arthritis and Immunology Research Program, Microarray Core Facility, The University Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA.
| | | | | | | | | |
Collapse
|
49
|
Categorically distinct acute stressors elicit dissimilar transcriptional profiles in the paraventricular nucleus of the hypothalamus. J Neurosci 2003. [PMID: 12843263 DOI: 10.1523/jneurosci.23-13-05607.2003] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The paraventricular hypothalamic nucleus (PVH) is a key site for integrating neuroendocrine, autonomic, and behavioral adjustments to diverse homeostatic challenges, including "physiological" (e.g., infection or hemorrhage) and "emotional" [e.g., restraint (RST) or footshock] stresses. Both types of challenges ultimately converge to activate common response systems represented in PVH, including the hypothalamo-pituitary-adrenal axis and the sympathoadrenal system. Oligonucleotide microarrays (U74A; Affymetrix, Santa Clara, CA) were used to compare and contrast gene expression profiles in the PVH elicited at 1 and 3 hr after acute exposure to representative physiological [intraperitoneal injection of 10 microg lipopolysaccharide (LPS)] and emotional (30 min RST) stressors. In general, the two challenges recruited relatively few genes in common, with the degree of overlap varying across functional classes of genes. The greatest degree of commonality was seen among signaling molecules and neuropeptides, whereas transcription factors upregulated by RST and LPS were largely distinct. Unexpectedly, RST induced a number of immune-related molecules, which were not regulated by LPS. Hybridization histochemical analyses localized a subset of responsive transcripts to the PVH and/or immediately adjoining regions. Immunerelated molecules in particular distributed broadly to vascular and other barrier-associated cell types. These global transcriptional profiles inform the search for early (transcription factors) and late (target genes) mechanisms in the modulation of PVH, and generalized CNS, responses to categorically distinct stressors.
Collapse
|
50
|
Abstract
Anaplastic large cell lymphoma (ALCL) is a neoplasm of activated lymphocytes, commonly expressing T-cell antigens and cytotoxic proteins. Histopathology reveals distinctive infiltration of sinuses and paracortical T-cell-rich regions of lymph nodes by tumor cells which have abundant cytoplasm and large irregular/convoluted nuclei, and which are frequently multinucleated with prominent nucleoli. ALCL often presents in advanced clinical stages with B symptoms; extranodal disease occurs in 40% of patients. The pathogenesis of systemic ALCL is linked to phosphorylation of a tyrosine kinase (ALK) resulting in unregulated growth of affected lymphoid cells. ALK is activated through chromosomal translocations/inversions with any of several partner genes, most commonly nucleophosmin (NPM). Downstream signal transduction pathway(s) are not fully defined but appear to involve phospholipase Cgamma, phosphatidylinositol (PI)3K/Akt, and STAT-3 and STAT-5 proteins. Primary cutaneous ALCL appears to have a different pathogenesis and better prognosis than does systemic ALCL, presenting as one or more skin tumors, usually localized. Excision or local irradiation is usually effective treatment. A clinically benign variant of primary cutaneous ALCL is lymphomatoid papulosis (LyP), characterized by recurrent crops of papules/nodules up to 2 cm in diameter which undergo spontaneous regression. LyP is managed by observation, ultraviolet light therapy, or low-dose methotrexate. LyP patients have a predisposition to develop malignant lymphomas, including Hodgkin's lymphoma, mycosis fungoides, and non-Hodgkin's lymphoma, by as yet unknown mechanisms. The prognosis for patients with LyP is otherwise excellent.
Collapse
MESH Headings
- Anaplastic Lymphoma Kinase
- Humans
- Lymphoma, Large-Cell, Anaplastic/classification
- Lymphoma, Large-Cell, Anaplastic/etiology
- Lymphoma, Large-Cell, Anaplastic/pathology
- Lymphoma, T-Cell, Cutaneous/classification
- Lymphoma, T-Cell, Cutaneous/etiology
- Lymphoma, T-Cell, Cutaneous/pathology
- Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases
- Signal Transduction
Collapse
Affiliation(s)
- Marshall E Kadin
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | |
Collapse
|