1
|
Santana-Gonçalves M, De Santis PB, Malmegrim KCR, Oliveira MC. T-cell Recovery After Autologous Hematopoietic Stem Cell Transplantation in Autoimmune Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:301-323. [PMID: 40067593 DOI: 10.1007/978-3-031-77921-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Over the last three decades, autologous hematopoietic stem cell transplantation (AHSCT) has emerged as a significant therapeutic strategy for patients with various refractory autoimmune diseases. Globally, more than 3000 AHSCT procedures have been performed for severe autoimmune diseases. The rationale behind this treatment is abrogation of autoreactivity; renovation of the immune system from the infused hematopoietic stem cells and establishment of a balanced, long-lasting, and self-tolerant immune system. Thymic rebound is a central event in the mechanisms of action of transplantation, promoting the generation of a new repertoire of T cells and reinstating self-tolerance. Here, we critically review the immunological mechanisms of AHSCT in various autoimmune disease settings, with a central focus on thymic rejuvenation. We understand that elucidating the mechanisms of action of the transplant and conducting immunological monitoring studies are crucial for comprehending the risks, benefits, and long-term efficacy of the treatment, thereby promoting functional improvements in patients with autoimmune diseases refractory to conventional treatment.
Collapse
Affiliation(s)
| | | | - Kelen C R Malmegrim
- Center for Cell-Based Therapy, Regional Blood Center, Ribeirão Preto, SP, Brazil.
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Maria Carolina Oliveira
- Center for Cell-Based Therapy, Regional Blood Center, Ribeirão Preto, SP, Brazil.
- Department of Internal Medicine (Clínica Médica), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
2
|
Quiros-Roldan E, Sottini A, Natali PG, Imberti L. The Impact of Immune System Aging on Infectious Diseases. Microorganisms 2024; 12:775. [PMID: 38674719 PMCID: PMC11051847 DOI: 10.3390/microorganisms12040775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Immune system aging is becoming a field of increasing public health interest because of prolonged life expectancy, which is not paralleled by an increase in health expectancy. As age progresses, innate and adaptive immune systems undergo changes, which are defined, respectively, as inflammaging and immune senescence. A wealth of available data demonstrates that these two conditions are closely linked, leading to a greater vulnerability of elderly subjects to viral, bacterial, and opportunistic infections as well as lower post-vaccination protection. To face this novel scenario, an in-depth assessment of the immune players involved in this changing epidemiology is demanded regarding the individual and concerted involvement of immune cells and mediators within endogenous and exogenous factors and co-morbidities. This review provides an overall updated description of the changes affecting the aging immune system, which may be of help in understanding the underlying mechanisms associated with the main age-associated infectious diseases.
Collapse
Affiliation(s)
- Eugenia Quiros-Roldan
- Department of Infectious and Tropical Diseases, ASST- Spedali Civili and DSCS- University of Brescia, 25123 Brescia, Italy;
| | - Alessandra Sottini
- Clinical Chemistry Laboratory, Services Department, ASST Spedali Civili of Brescia, 25123 Brescia, Italy;
| | - Pier Giorgio Natali
- Mediterranean Task Force for Cancer Control (MTCC), Via Pizzo Bernina, 14, 00141 Rome, Italy;
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, P. le Spedali Civili, 1, 25123 Brescia, Italy
| |
Collapse
|
3
|
Chang H, Marquez J, Chen BK, Kim DM, Cheng ML, Liu EV, Yang H, Zhang L, Sinha M, Cheung A, Kwek SS, Chow ED, Bridge M, Aggarwal RR, Friedlander TW, Small EJ, Anderson M, Fong L. Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer. Cancer Immunol Res 2024; 12:453-461. [PMID: 38276989 PMCID: PMC10993769 DOI: 10.1158/2326-6066.cir-23-0184] [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: 02/27/2023] [Revised: 09/21/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024]
Abstract
Denosumab is a fully human mAb that binds receptor activator of NFκB ligand (RANKL). It is routinely administered to patients with cancer to reduce the incidence of new bone metastasis. RANK-RANKL interactions regulate bone turnover by controlling osteoclast recruitment, development, and activity. However, these interactions also can regulate immune cells including dendritic cells and medullary thymic epithelial cells. Inhibition of the latter results in reduced thymic negative selection of T cells and could enhance the generation of tumor-specific T cells. We examined whether administering denosumab could modify modulate circulating immune cells in patients with cancer. Blood was collected from 23 patients with prostate cancer and 3 patients with renal cell carcinoma, all of whom had advanced disease and were receiving denosumab, prior to and during denosumab treatment. Using high-dimensional mass cytometry, we found that denosumab treatment by itself induced modest effects on circulating immune cell frequency and activation. We also found minimal changes in the circulating T-cell repertoire and the frequency of new thymic emigrants with denosumab treatment. However, when we stratified patients by whether they were receiving chemotherapy and/or steroids, patients receiving these concomitant treatments showed significantly greater immune modulation, including an increase in the frequency of natural killer cells early and classical monocytes later. We also saw broad induction of CTLA-4 and TIM3 expression in circulating lymphocytes and some monocyte populations. These findings suggest that denosumab treatment by itself has modest immunomodulatory effects, but when combined with conventional cancer treatments, can lead to the induction of immunologic checkpoints. See related Spotlight by Nasrollahi and Davar, p. 383.
Collapse
Affiliation(s)
- Hewitt Chang
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Jaqueline Marquez
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Brandon K. Chen
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Daniel M. Kim
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Michael L. Cheng
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Eric V. Liu
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Hai Yang
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - Li Zhang
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - Meenal Sinha
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Alexander Cheung
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Serena S. Kwek
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Eric D. Chow
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
- Department of Biochemistry and Biophysics, Center for Advanced Technologies, University of California San Francisco, San Francisco, California
| | - Mark Bridge
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Rahul R. Aggarwal
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Terence W. Friedlander
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Eric J. Small
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Mark Anderson
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| |
Collapse
|
4
|
Zhao Q, Dai R, Li Y, Wang Y, Chen X, Shu Z, Zhou L, Ding Y, Tang X, Zhao X. Trends in TREC values according to age and gender in Chinese children and their clinical applications. Eur J Pediatr 2022; 181:529-538. [PMID: 34405301 DOI: 10.1007/s00431-021-04223-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/28/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
T cell receptor excision circles (TRECs) are small circularized DNA elements produced during rearrangement of T cell receptor (TCR) genes. Because TRECs are fairly stable, do not replicate during mitosis, and are not diluted during division of naïve T cells (Dion et al. [1]), they are suitable for assessing the number of newly formed T cells (Ping and Denise [2]). In this study, we detected TRECs in 521 healthy Chinese children aged 0-18 years in different clinical settings. The TRECs decrease with aging and show lower levels in preterm and low birth weight (BW) babies compared to those in full-term infants, while the preterm babies can also show comparable levels of TRECs when they have a gestation age (GA)-matched BW. We found a strong correlation between TRECs and peripheral CD4 naïve T cell numbers, which was age-related. We also analyzed the TRECs in different PIDs. Since T cell defects vary in PIDs, TREC levels change inconsistently. For example, in Wiskott-Aldrich syndrome (WAS), combining the level of TREC with lymphocyte subsets can help to distinguish subtypes of disease.Conclusion: We established the reference value range for TRECs by evaluating children below 18 years old in China, which could be used to screen for PIDs during early life. What is Known: • The TREC levels are decreased with age, and there is a positive correlation between TRECs and the numbers of naïve T cells. What is New: • This is the largest study to determine TREC reference levels in healthy Chinese pediatric, we provide solid data showing a correlation between CD4 naïve T cell counts and TREC levels according to age. We point out the GA matched BW is need to be considered during the SCID newborn screening. We are the first group showed that TREC levels can help clinician distinguish different WAS phenotype.
Collapse
Affiliation(s)
- Qin Zhao
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Rongxin Dai
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong District, Chongqing, 400014, China
| | - Yanan Li
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yanping Wang
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Xuemei Chen
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Zhou Shu
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong District, Chongqing, 400014, China
| | - Lina Zhou
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yuan Ding
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Department of Health Management, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Xuemei Tang
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong District, Chongqing, 400014, China
| | - Xiaodong Zhao
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China. .,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China. .,Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong District, Chongqing, 400014, China.
| |
Collapse
|
5
|
Kwok JSY, Cheung SKF, Ho JCY, Tang IWH, Chu PWK, Leung EYS, Lee PPW, Cheuk DKL, Lee V, Ip P, Lau YL. Establishing Simultaneous T Cell Receptor Excision Circles (TREC) and K-Deleting Recombination Excision Circles (KREC) Quantification Assays and Laboratory Reference Intervals in Healthy Individuals of Different Age Groups in Hong Kong. Front Immunol 2020; 11:1411. [PMID: 32765500 PMCID: PMC7378446 DOI: 10.3389/fimmu.2020.01411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/02/2020] [Indexed: 01/10/2023] Open
Abstract
The clinical experience gathered throughout the years has raised awareness of primary immunodeficiency diseases (PIDD). T cell receptor excision circles (TREC) and kappa-deleting recombination excision circles (KREC) assays for thymic and bone marrow outputs measurement have been widely implemented in newborn screening (NBS) programs for Severe Combined Immunodeficiency. The potential applications of combined TREC and KREC assay in PIDD diagnosis and immune reconstitution monitoring in non-neonatal patients have been suggested. Given that ethnicity, gender, and age can contribute to variations in immunity, defining the reference intervals of TREC and KREC levels in the local population is crucial for setting up cut-offs for PIDD diagnosis. In this retrospective study, 479 healthy Chinese sibling donors (240 males and 239 females; age range: 1 month-74 years) from Hong Kong were tested for TREC and KREC levels using a simultaneous quantitative real-time PCR assay. Age-specific 5th-95th percentile reference intervals of TREC and KREC levels (expressed in copies per μL blood and copies per 106 cells) were established in both pediatric and adult age groups. Significant inverse correlations between age and both TREC and KREC levels were observed in the pediatric age group. A significant higher KREC level was observed in females than males after 9-12 years of age but not for TREC. Low TREC or KREC levels were detected in patients diagnosed with mild or severe PIDD. This assay with the established local reference intervals would allow accurate diagnosis of PIDD, and potentially monitoring immune reconstitution following haematopoietic stem cell transplantation or highly active anti-retroviral therapy in the future.
Collapse
Affiliation(s)
- Janette S. Y. Kwok
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong, Hong Kong
| | - Stephen K. F. Cheung
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong, Hong Kong
| | - Jenny C. Y. Ho
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong, Hong Kong
| | - Ivan W. H. Tang
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong, Hong Kong
| | - Patrick W. K. Chu
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong, Hong Kong
| | - Eric Y. S. Leung
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong, Hong Kong
| | - Pamela P. W. Lee
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Daniel K. L. Cheuk
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Vincent Lee
- Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Patrick Ip
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Y. L. Lau
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
6
|
da Rocha LKA, Freschi de Barros S, Bandeira F, Bollini A, Testa LHDA, Simione AJ, Souza MDOE, Zanetti LP, de Oliveira LCS, Dos Santos ACF, de Souza MP, Colturado VAR, Kalil J, Machado CM, Guilherme L. Thymopoiesis in Pre- and Post-Hematopoietic Stem Cell Transplantation. Front Immunol 2018; 9:1889. [PMID: 30245685 PMCID: PMC6137257 DOI: 10.3389/fimmu.2018.01889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/31/2018] [Indexed: 12/20/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is an important therapeutic option for some hematological diseases. However, patients who undergo HSCT acquire a state of immunodeficiency that causes significant mortality. Reconstitution of thymic function is needed to support the immune system. One way to measure thymic function is through T-cell receptor excision circle (TREC) quantification. TRECs are generated by T-cell receptor gene rearrangements during T-cell maturation in the thymus and represent a reliable marker for thymic output. In this study, we aimed to assess aging and malignant hematological diseases as two important factors that may influence thymic output before HSCT. We observed that patients before HSCT presented signal joint TREC (sjTREC) numbers lower than 606.55 copies/μg DNA (low values) compared with healthy individuals, with an odds ratio (OR) of 12.88 [95% confidence interval (CI): 5.26–31.53; p < 0.001]. Our results showed that a group of older individuals (≥50 years old), comprising both healthy individuals and patients, had an OR of 10.07 (95% CI: 2.80–36.20) for low sjTREC values compared with younger individuals (≤24 years old; p < 0.001). Multiple logistic regression analysis confirmed that both older age (≥50 years old) and malignant hematological diseases and their treatments were important and independent risk factors related to thymic function impairment (p < 0.001). The median sjTREC value for patients of all ages was significantly lower than the sjTREC median for the subgroup of older healthy individuals (≥50 years old; p < 0.001). These data suggested that patients before HSCT and healthy individuals exhibited age-dependent thymic impairment, and that prior treatment for hematological diseases may exacerbate aging-related deterioration of natural thymic function. Furthermore, we analyzed these patients 9 months post-HSCT and compared patients who underwent autologous HSCT with those who underwent allogeneic HSCT. Both groups of patients achieved sjTREC copy numbers similar to those of healthy individuals. We did not find a close relationship between impaired thymic function prior to HSCT and worse thymic recovery after HSCT.
Collapse
Affiliation(s)
- Luis Klaus A da Rocha
- Laboratory of Immunology, Heart Institute (InCor), Clinical Hospital, University of São Paulo, São Paulo, Brazil
| | - Samar Freschi de Barros
- Laboratory of Immunology, Heart Institute (InCor), Clinical Hospital, University of São Paulo, São Paulo, Brazil
| | - Francine Bandeira
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | - Alexia Bollini
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | | | - Anderson João Simione
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | - Marina de O E Souza
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | - Lilian P Zanetti
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | | | | | - Mair Pedro de Souza
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | | | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (InCor), Clinical Hospital, University of São Paulo, São Paulo, Brazil
| | - Clarisse M Machado
- Laboratory of Virology, Tropical Medicine Institute (IMT), University of São Paulo, São Paulo, Brazil
| | - Luiza Guilherme
- Laboratory of Immunology, Heart Institute (InCor), Clinical Hospital, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Silva SL, Sousa AE. Establishment and Maintenance of the Human Naïve CD4 + T-Cell Compartment. Front Pediatr 2016; 4:119. [PMID: 27843891 PMCID: PMC5086629 DOI: 10.3389/fped.2016.00119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/17/2016] [Indexed: 12/26/2022] Open
Abstract
The naïve CD4+ T-cell compartment is considered essential to guarantee immune competence throughout life. Its replenishment with naïve cells with broad diverse receptor repertoire, albeit with reduced self-reactivity, is ensured by the thymus. Nevertheless, cumulative data support a major requirement of post-thymic proliferation both for the establishment of the human peripheral naïve compartment during the accelerated somatic growth of childhood, as well as for its lifelong maintenance. Additionally, a dynamic equilibrium is operating at the cell level to fine-tune the T-cell receptor threshold to activation and survival cues, in order to counteract the continuous naïve cell loss by death or conversion into memory/effector cells. The main players in these processes are low-affinity self-peptide/MHC and cytokines, particularly IL-7. Moreover, although naïve CD4+ T-cells are usually seen as a homogeneous population regarding stage of maturation and cell differentiation, increasing evidence points to a variety of phenotypic and functional subsets with distinct homeostatic requirements. The paradigm of cells committed to a distinct lineage in the thymus are the naïve regulatory T-cells, but other functional subpopulations have been identified based on their time span after thymic egress, phenotypic markers, such as CD31, or cytokine production, namely IL-8. Understanding the regulation of these processes is of utmost importance to promote immune reconstitution in several clinical settings, namely transplantation, persistent infections, and aging. In this mini review, we provide an overview of the mechanisms underlying human naïve CD4+ T-cell homeostasis, combining clinical data, experimental studies, and modeling approaches.
Collapse
Affiliation(s)
- Susana L Silva
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Centro de Imunodeficiências Primárias, Lisboa, Portugal; Clinica Universitária de Imunoalergologia, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisboa, Portugal
| | - Ana E Sousa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Centro de Imunodeficiências Primárias, Lisboa, Portugal
| |
Collapse
|
8
|
Duggleby RC, Madrigal JA. Methods of detection of immune reconstitution and T regulatory cells by flow cytometry. Methods Mol Biol 2014; 1109:159-86. [PMID: 24473784 DOI: 10.1007/978-1-4614-9437-9_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Allogeneic hematopoietic stem cell therapy (HSCT) remains one of the few curative treatments for high-risk hematological malignancies (high-risk leukemia, myelodysplastic syndromes, advanced myeloproliferative disorders, high-risk lymphomas, and multiple myeloma) and is currently applied in more than 15,000 patients per year in Europe. Following HSCT, patients experience a period of reconstitution of the immune system, which seems to be highly dependent on conditioning, immunosuppression regimes, and the level of adverse events the patients experience. During this reconstitution period, the patient is immune compromised and susceptible to opportunistic infections and disease relapse. Consequently, a large number of clinical studies have been devoted to monitoring the recovery of the immune system following HSCT in the hopes of determining which cellular subsets are indicative of a favorable outcome. In this chapter we review the methods that have been employed to monitor the immune reconstitution and what clinical observations have been made. Of particular interest is the regulatory T cell (Treg) subset, which has been associated with tolerance and has been the subject of recent clinical trials as a possible cellular therapy for rejection reactions. Finally we will detail a proposed methodology for the flow cytometric assessment of cellular reconstitution post-HSCT.
Collapse
|
9
|
Arismendi MI, Kallás EG, Santos BAND, Carneiro-Sampaio MMS, Kayser C. Thymopoiesis and regulatory T cells in healthy children and adolescents. Clinics (Sao Paulo) 2012; 67:425-9. [PMID: 22666784 PMCID: PMC3351266 DOI: 10.6061/clinics/2012(05)04] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 01/11/2012] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES The purpose of this study was to investigate the association between T cell receptor excision circle levels in peripheral blood mononuclear cells and regulatory T cells that co-express CD25 and Foxp3 in healthy children and adolescents of different ages. MATERIALS AND METHODS The quantification of signal-joint T-cell receptor excision circle levels in the genomic DNA of peripheral blood mononuclear cells was performed using real-time quantitative PCR. The analysis of CD4, CD8, CD25, and Foxp3 expression was performed using flow cytometry. RESULTS Ninety-five healthy controls (46 females and 49 males) ranging in age from 1 to 18 years were analyzed. The mean T-cell receptor excision circle count in all individuals was 89.095 ± 36.790 T-cell receptor excision circles per microgram of DNA. There was an inverse correlation between T-cell receptor excision circles counts and age (r = -0.846; p<0.001) as well as between the proportion of CD4(+)CD25(+)Foxp3(+) T cells and age (r = -0.467; p = 0.04). In addition, we observed a positive correlation between the amount of CD4(+)CD25(+)Foxp3(+) T cells and the amount of T-cell receptor excision circles per microgram of DNA in individuals of all ages (r = -0.529; p = 0.02). CONCLUSIONS In this study, we observed a decrease in the thymic function with age based on the fact that the level of T-cell receptor excision circles in the peripheral blood positively correlated with the proportion of regulatory T cells in healthy children and adolescents. These findings indicate that although T-cell receptor excision circles and regulatory T cells levels decrease with age, homeostasis of the immune system and relative regulatory T cells population levels are maintained in the peripheral blood.
Collapse
Affiliation(s)
- Maria Izabel Arismendi
- Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, Departamento de Pediatria, Instituto da Criança, Brasil
| | | | | | | | | |
Collapse
|
10
|
Lima K, Abrahamsen TG, Foelling I, Natvig S, Ryder LP, Olaussen RW. Low thymic output in the 22q11.2 deletion syndrome measured by CCR9+CD45RA+ T cell counts and T cell receptor rearrangement excision circles. Clin Exp Immunol 2010; 161:98-107. [PMID: 20491792 DOI: 10.1111/j.1365-2249.2010.04152.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Thymic hypoplasia is a frequent feature of the 22q11.2 deletion syndrome, but we know little about patients' age-related thymic output and long-term consequences for their immune system. We measured the expression of T cell receptor rearrangement excision circles (TREC) and used flow cytometry for direct subtyping of recent thymic emigrant (RTE)-related T cells in 43 patients (aged 1-54 years; median 9 years) from all over Norway and in age-matched healthy controls. Thymic volumes were estimated by ultrasound in patients. TREC levels correlated well with RTE-related T cells defined by co-expression of CD3, CD45RA and CCR9 (r=0.84) as well as with the CD4+ and CD8+ T cell subtypes. RTE-related T cell counts also paralleled age-related TREC reductions. CD45RA+ T cells correlated well with absolute counts of CD4+ (r=0.87) and CD8+ (r=0.75) RTE-related T cells. Apart from CD45RA- T cells, all T cell subsets were lower in patients than in controls. Thymic volumes correlated better with RTE-related cells (r=0.46) than with TREC levels (r=0.38). RTE-related T cells and TREC levels also correlated well (r=0.88) in patients without an identifiable thymus. Production of RTEs is impaired in patients with a 22q11.2 deletion, and CCR9 appears to be a good marker for RTE-related T cells.
Collapse
Affiliation(s)
- K Lima
- Section of Endocrinology, Faculty Division Akershus University Hospital, University of Oslo, Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
11
|
Zlamy M, Prelog M. Thymectomy in early childhood: a model for premature T cell immunosenescence? Rejuvenation Res 2010; 12:249-58. [PMID: 19673593 DOI: 10.1089/rej.2009.0864] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The thymus is the main source of recent thymic emigrants (RTE) and naïve T cells. The aging of the immune system (immunosenescence) is characterized by loss of thymic function, decreased numbers of RTE, peripheral proliferation of mature T cells, and oligoclonal expansions of specific T cell subpopulations. As shown in several studies, thymectomized patients demonstrate signs of premature immunosenescence reminiscent of aged people, such as decreased proportions of naïve T cells and RTE, a compensatory increase of mature T cell subpopulations with increased proliferation rates, restriction of the T cell receptor repertoire, and a delayed response to new antigens and vaccinations. This review demonstrates that, despite some limitations, childhood thymectomy may serve as an useful model for premature immunosenescence, mimicking changes expected after physiological thymus involution in the elderly. Thus, it may prove an insightful tool for obtaining better understanding of human naïve T cell development, thymic function, and maintenance of the naïve T cell pool.
Collapse
Affiliation(s)
- Manuela Zlamy
- Department of Pediatrics, Pediatrics I, Medical University Innsbruck, Austria
| | | |
Collapse
|
12
|
van Dommelen SLH, Rizzitelli A, Chidgey A, Boyd R, Shortman K, Wu L. Regeneration of dendritic cells in aged mice. Cell Mol Immunol 2010; 7:108-15. [PMID: 20118970 DOI: 10.1038/cmi.2009.114] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Age-related thymic involution causes a decreased output of thymocytes from the thymus, thereby resulting in impairment of T cell-mediated immunity. While alterations in the T cell and non-haematopoietic stromal compartments have been described, the effects of thymic involution on thymic dendritic cells (DC) are not clearly known. Thymic DC play an essential role in shaping T cell-mediated immune responses by deleting self-reactive thymocytes to establish central tolerance and by inducing regulatory T-cell (Treg) development. It is therefore important to assess the prevalence of and alterations to thymic DC with age, as this may impact on their function. We assessed the numbers and proportions of the three distinct subsets of thymic DC in ageing mice, and showed that these subsets are differentially regulated. This is expected as thymic DC subsets have different origins of development. We further assessed the responses of thymic DC in a regenerative environment, such as that induced by sex-steroid ablation (SSA), and clearly showed that, consistent with global thymus regrowth, all three DC populations increased in numbers and regained their relative proportions to thymocytes after an initial lag period. These findings are important for the clinical translation of thymic regenerative approaches, and indicate that SSA facilitates the maintenance of critical processes such as negative selection and Treg induction through promoting thymic DC regeneration.
Collapse
|
13
|
Li B, Li YQ, Yang LJ, Chen SH, Yu W, Chen JY, Liu WW. Decreased T-cell receptor excision DNA circles in peripheral blood mononuclear cells among benzene-exposed workers. Int J Immunogenet 2009; 36:107-11. [PMID: 19228219 DOI: 10.1111/j.1744-313x.2009.00832.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Benzene is a volatile aromatic hydrocarbon solvent which is widely used in many industries. The chronic exposure of humans to benzene in the workplace has been associated with blood disorders, as well as toxicity in lymphopoiesis, including aplastic anaemia and leukaemia. However, the mechanisms of benzene-induced haematotoxicity and leukaemogenesis remain unclear. In this study, we investigated the level of T-cell receptor excision DNA circles (TRECs) in peripheral blood mononuclear cells (PBMCs) in benzene-exposed workers. This would therefore be considered as a potential marker for estimates of thymic output and an evaluation of the content of naïve T-cells. It is hoped that the data will bring a comprehensive understanding on the influence of benzene exposure in the host T-cell immune function. Quantitative detection of TRECs in DNA of PBMCs from benzene-exposed workers was preformed by real-time polymerase chain reaction using the TaqMan technique. The benzene-exposed workers were divided into four groups, and 27 normal individuals were served as controls. The result indicated that the TRECs levels of all benzene-exposed groups were significantly decreased as compared with those of controls. In conclusion, the recent thymic output function and the T-cell immune function were apparently impaired in workers after benzene exposure.
Collapse
Affiliation(s)
- B Li
- Institute of Hematology, Medical College, Jinan University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Ogle BM, Knudsen BE, Nishitai R, Ogata K, Platt JL. Toward development and production of human T cells in swine for potential use in adoptive T cell immunotherapy. Tissue Eng Part A 2009; 15:1031-40. [PMID: 18826341 PMCID: PMC2810409 DOI: 10.1089/ten.tea.2008.0117] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Accepted: 07/08/2008] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy and vaccination for cancer or infection are generally approached by administration of antigen or stimulation of antigen-presenting cells or both. These measures may fail if the treated individual lacks T cells specific for the immunogen(s). We tested another strategy-the generation of new T cells from hematopoietic stem cells that might be used for adoptive immunotherapy. To test this concept, we introduced T cell-depleted human bone marrow cells into fetal swine and tested the swine for human T cells at various times after birth. Human T cells were detected in the thymus and blood of the treated swine. These cells were generated de novo as they contained human T cell receptor excision circles not present in the T cell-depleted bone marrow. The human T cells were highly diverse and included novel specificities capable of responding to antigen presented by human antigen-presenting cells. Our findings constitute a first step in a new promising approach to immunotherapy in which tumor- or virus-specific T cell clones lacking in an individual might be generated in a surrogate host from hematopoietic stem cells of the individual to be treated.
Collapse
Affiliation(s)
- Brenda M. Ogle
- Transplantation Biology Program, Mayo Clinic, Rochester, Minnesota
- Department of Physiology, Mayo Clinic, Rochester, Minnesota
| | - Bruce E. Knudsen
- Transplantation Biology Program, Mayo Clinic, Rochester, Minnesota
| | - Ryuta Nishitai
- Department of Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Kiyoshi Ogata
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Jeffrey L. Platt
- Transplantation Biology Program, Mayo Clinic, Rochester, Minnesota
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
- Department of Pediatrics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
15
|
Krenger W, Holländer GA. The immunopathology of thymic GVHD. Semin Immunopathol 2008; 30:439-56. [PMID: 18974988 DOI: 10.1007/s00281-008-0131-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 09/30/2008] [Indexed: 12/11/2022]
Abstract
The clinical success of allogeneic hematopoietic stem cell transplantation (HSCT) depends on the appropriate reconstitution of the host's immune system. While recovery of T-cell immunity may occur in transplant recipients via both thymus-dependent and thymus-independent pathways, the regeneration of a population of phenotypically naive T cells with a broad receptor repertoire relies entirely on the de novo generation of T-cells in the thymus. Preclinical models and clinical studies of allogeneic HSCT have identified the thymus as a target of graft-versus-host disease (GVHD), thus limiting T-cell regeneration. The present review focuses on recent insight into how GVHD affects thymic structure and function and how this knowledge may aid in the design of new strategies to improve T-cell reconstitution following allogeneic HSCT.
Collapse
Affiliation(s)
- Werner Krenger
- Department of Biomedicine, University of Basel, Basel, 4005, Switzerland.
| | | |
Collapse
|
16
|
Prelog M, Keller M, Geiger R, Brandstätter A, Würzner R, Schweigmann U, Zlamy M, Zimmerhackl LB, Grubeck-Loebenstein B. Thymectomy in early childhood: significant alterations of the CD4(+)CD45RA(+)CD62L(+) T cell compartment in later life. Clin Immunol 2008; 130:123-32. [PMID: 18977182 DOI: 10.1016/j.clim.2008.08.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 08/13/2008] [Accepted: 08/21/2008] [Indexed: 01/01/2023]
Abstract
The study was aimed to assess indicators of immunosenescence, such as the total counts of peripheral blood CD4(+)CD45RA(+)CD62L(+) (naive) T cells, the numbers of T cell receptor excision circles (TRECs), and Ki67-expression as marker of peripheral replication in thymectomized patients (TP) (n=101) compared to age-matched healthy donors (HD) (n=81). In TP, there was an inverse correlation between naive T cells and chronological age (p<0.001) or time post thymectomy (p<0.001). TP demonstrated lower TREC numbers in naive T cells compared to HD (p<0.001). TREC numbers negatively correlated with time post thymectomy (p<0.001). Percentages of Ki67-expresssing naive T cells were higher in TP compared to HD (p<0.05). The findings of the presented long-term follow up cohort of thymectomized patients indicate that changes of the peripheral naive T cell subset in TP may resemble the findings of an aging immune system in elderly persons after thymic involution. Our data provide evidence that peripheral T cell homeostasis in TP is maintained at minimal levels mainly by extrathymic expansion of existing naive T cells in the periphery to compensate the diminished thymic output.
Collapse
Affiliation(s)
- Martina Prelog
- Department of Pediatrics, Pediatrics I, Medical University Innsbruck, Anichstr. 35 A-6020 Innsbruck, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Huang X, Moore DJ, Ketchum RJ, Nunemaker CS, Kovatchev B, McCall AL, Brayman KL. Resolving the conundrum of islet transplantation by linking metabolic dysregulation, inflammation, and immune regulation. Endocr Rev 2008; 29:603-30. [PMID: 18664617 PMCID: PMC2819735 DOI: 10.1210/er.2008-0006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Accepted: 05/29/2008] [Indexed: 02/08/2023]
Abstract
Although type 1 diabetes cannot be prevented or reversed, replacement of insulin production by transplantation of the pancreas or pancreatic islets represents a definitive solution. At present, transplantation can restore euglycemia, but this restoration is short-lived, requires islets from multiple donors, and necessitates lifelong immunosuppression. An emerging paradigm in transplantation and autoimmunity indicates that systemic inflammation contributes to tissue injury while disrupting immune tolerance. We identify multiple barriers to successful islet transplantation, each of which either contributes to the inflammatory state or is augmented by it. To optimize islet transplantation for diabetes reversal, we suggest that targeting these interacting barriers and the accompanying inflammation may represent an improved approach to achieve successful clinical islet transplantation by enhancing islet survival, regeneration or neogenesis potential, and tolerance induction. Overall, we consider the proinflammatory effects of important technical, immunological, and metabolic barriers including: 1) islet isolation and transplantation, including selection of implantation site; 2) recurrent autoimmunity, alloimmune rejection, and unique features of the autoimmune-prone immune system; and 3) the deranged metabolism of the islet transplant recipient. Consideration of these themes reveals that each is interrelated to and exacerbated by the other and that this connection is mediated by a systemic inflammatory state. This inflammatory state may form the central barrier to successful islet transplantation. Overall, there remains substantial promise in islet transplantation with several avenues of ongoing promising research. This review focuses on interactions between the technical, immunological, and metabolic barriers that must be overcome to optimize the success of this important therapeutic approach.
Collapse
Affiliation(s)
- Xiaolun Huang
- Department of Surgery, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Immunologic defects in 22q11.2 deletion syndrome. J Allergy Clin Immunol 2008; 122:362-7, 367.e1-4. [PMID: 18485468 DOI: 10.1016/j.jaci.2008.03.033] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Revised: 03/28/2008] [Accepted: 03/28/2008] [Indexed: 01/07/2023]
Abstract
BACKGROUND 22q11.2 Deletion syndrome, the most common congenital chromosome deletion syndrome, is associated with developmental defects including cardiac abnormalities and hypoplasia or abnormal migration of the thymus. These patients have variable defects in T-cell immunity with an increased incidence of infection and autoimmune disease. OBJECTIVE To investigate the immunologic constitution of children with 22q11.2 deletion syndrome. METHODS We characterized the immunologic constitution of 27 children with 22q11.2 deletion syndrome and 54 healthy controls by flow-cytometric analysis of peripheral blood lymphocyte populations. RESULTS Patients exhibited decreased T-cell numbers, although the normal age-related decrease in T-cell numbers was slower than in healthy children. There was a significant decrease in FoxP3(+) natural regulatory T (nTreg) cells with a strong correlation between nTreg cells and recent T-cell emigrants from the thymus, suggesting a link between the nTreg cell population and thymic function. Although total B-cell numbers were unaffected, patients showed a significantly decreased proportion of memory B cells in the B-cell pool. CONCLUSION Lower nTreg cells in patients suggest that the generation and maintenance of these cells in children is related to thymic function. In addition to T-cell abnormalities classically seen in this syndrome, subtle defects in the B-cell compartment may also be seen.
Collapse
|
19
|
Gravekamp C, Kim SH, Castro F. Cancer vaccination: manipulation of immune responses at old age. Mech Ageing Dev 2008; 130:67-75. [PMID: 18561984 DOI: 10.1016/j.mad.2008.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 04/18/2008] [Accepted: 05/02/2008] [Indexed: 01/07/2023]
Abstract
The incidence of cancer has increased over the last decade, mainly due to an increase in the elderly population. Vaccine therapy for cancer is less toxic than chemotherapy or radiation and could be, therefore, especially effective in older, more frail cancer patients. However, it has been shown that older individuals do not respond to vaccine therapy as well as younger adults. This has been attributed to T-cell unresponsiveness, a phenomenon also observed in cancer patients per se. This review summarizes the current knowledge of impaired T-cell responses in cancer patients and the elderly, and the results of cancer vaccination in preclinical models at young and old age. Finally, various approaches how to manipulate immune responses against cancer by vaccination at older age will be proposed.
Collapse
Affiliation(s)
- Claudia Gravekamp
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA.
| | | | | |
Collapse
|
20
|
Hince M, Sakkal S, Vlahos K, Dudakov J, Boyd R, Chidgey A. The role of sex steroids and gonadectomy in the control of thymic involution. Cell Immunol 2008; 252:122-38. [PMID: 18294626 DOI: 10.1016/j.cellimm.2007.10.007] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Accepted: 10/30/2007] [Indexed: 01/25/2023]
Abstract
A major underlying cause for aging of the immune system is the structural and functional atrophy of the thymus, and associated decline in T cell genesis. This loss of naïve T cells reduces adaptive immunity to new stimuli and precipitates a peripheral bias to memory cells against prior antigens. Whilst multiple mechanisms may contribute to this process, the temporal alliance of thymic decline with puberty has implicated a causative role for sex steroids. Accordingly ablation of sex steroids induces profound thymic rejuvenation. Although the thymus retains some, albeit highly limited, function in healthy adults, this is insufficient for resurrecting the T cell pool following cytoablative treatments such as chemo- and radiation-therapy and AIDS. Increased risk of opportunistic infections and cancer relapse or appearance, are a direct consequence. Temporary sex steroid ablation may thus provide a clinically effective means to regenerate the thymus and immune system in immunodeficiency states.
Collapse
Affiliation(s)
- Melanie Hince
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building-75, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | | | | | | | | | | |
Collapse
|
21
|
Thymopoiesis, regulatory T cells, and TCRVbeta expression in thymoma with and without myasthenia gravis, and modulatory effects of steroid therapy. J Clin Immunol 2007; 28:194-206. [PMID: 18000743 DOI: 10.1007/s10875-007-9147-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Accepted: 10/16/2007] [Indexed: 10/22/2022]
Abstract
We analyzed thymocyte and thymic regulatory T cell (CD4SPCD25+Foxp3+cells, Treg) development in thymoma with and without myasthenia gravis (MG, MG-thymoma, non-MG-thymoma) and in MG-associated non-neoplastic thymus (MG-NNT). An increased number of immature CD4+CD8(-)CD3(-) thymocytes through the CD4+CD8+ to CD4+CD8(-) transition and an abnormal T cell receptor Vbeta (TCRVbeta) development through the CD4+CD8+ to CD4(-)CD8+ transition were seen both in MG-and non-MG-thymomas. Terminal thymopoiesis, i.e., CD45RA+ cells within the CD4+CD8(-)CD3+ and CD8+CD4(-)CD3+ subsets, was skewed towards the CD4+ compartment in MG-thymoma and CD8+ compartment in non-MG-thymoma, but thymic export was increased only in the latter in keeping with the hypothesis that CD8+ lymphocytes may play a role in the initial stages of autosensitization and in disagreement with the relevance of an increased output of CD4+ T lymphocytes in paraneoplastic MG. Treg level in normal thymus and MG-NNT and both MG- and non-MG-thymoma was similar, and TCRVbeta development in Treg cells was slightly altered in thymoma but irrespective of MG presence. Thus, the relevance of a defective Treg development in MG context remains to be established. Most alterations in thymopoiesis were corrected by therapeutic corticosteroid administration, and the effects of steroid administration may be mediated by thymic microenvironment.
Collapse
|
22
|
Hudson LL, Markert ML, Devlin BH, Haynes BF, Sempowski GD. Human T cell reconstitution in DiGeorge syndrome and HIV-1 infection. Semin Immunol 2007; 19:297-309. [PMID: 18035553 PMCID: PMC2189557 DOI: 10.1016/j.smim.2007.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2007] [Accepted: 10/02/2007] [Indexed: 01/19/2023]
Abstract
The thymus is essential for proper development and maintenance of a broad T cell repertoire capable of recognizing a wide-range of foreign antigens. Recent advances in multicolor flow cytometry, non-invasive imaging techniques, and molecular assessments of thymic function have enabled a more comprehensive characterization of human thymic output in clinical settings than in the past. These techniques have been particularly valuable in monitoring human T cells after therapeutic thymic grafting for complete DiGeorge syndrome and during HIV-1 infection and AIDS. By defining the degree and mechanisms of T cell reconstitution in these settings, clinical investigators and primary caregivers have been able to better diagnose, treat and care for individuals with congenital or acquired immune deficiencies associated with loss of thymic function.
Collapse
Affiliation(s)
- Lori L. Hudson
- From the Duke University Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA 27710
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA 27710
| | - M. Louise Markert
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA 27710
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA 27710
| | - Blythe H. Devlin
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA 27710
| | - Barton F. Haynes
- From the Duke University Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA 27710
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA 27710
| | - Gregory D. Sempowski
- From the Duke University Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA 27710
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA 27710
| |
Collapse
|
23
|
Fu YW, Wu DP, Cen JN, Feng YF, Chang WR, Zhu ZL, Qiu QC, Zhu P. Patterns of T-cell reconstitution by assessment of T-cell receptor excision circle and T-cell receptor clonal repertoire after allogeneic hematopoietic stem cell transplantation in leukemia patients ? a study in Chinese patients. Eur J Haematol 2007; 79:138-45. [PMID: 17608713 DOI: 10.1111/j.1600-0609.2007.00885.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Successful allogeneic hematopoietic stem cell transplantation (HSCT) requires reconstitution normal T-cell immunity. Measurement of T-cell receptor excision circles (TRECs) and T-cell receptor beta (TCRBV) CDR3 repertoire is a means of quantifying recent thymic T-cell production and reflecting antigen-specific T-cell clones proliferation. METHODS We used real-time quantitative PCR to detect TRECs from 43 Chinese patients who underwent three kind of allo-HSCT without T-cell depletion. RT-PCR was performed to amplify 24 subfamily genes of TCRBV in 24 patients of them. RESULTS For haploidentical-D group, the TRECs numbers were lower up to 24 months. For matched-sibling donor (MSD) group, the recovery of TRECs was faster than those of other two groups. TRECs values in matched-unrelated donor (MUD) were in the middle. During 2-19 months after transplantation, there were 6-16 BV subfamilies expressed and 33-48% of them were polyclones. The usage rate of TCRBV and percentage of polyclones in haploidentical-D were less than those of other two groups. Twenty-three CDR3 molecules were obtained from nine patients who were potentially associated with GVHD or CMV infection. CONCLUSIONS Analyzing the changes of TCRBV repertoire and measuring TRECs during immune reconstitution would be useful to determine the host's current immune status and ability of T-cell immune reconstitution and also to find antigen-specific T-cell clones in the three kinds of HSCT.
Collapse
Affiliation(s)
- Yue Wen Fu
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Goldberg GL, Alpdogan O, Muriglan SJ, Hammett MV, Milton MK, Eng JM, Hubbard VM, Kochman A, Willis LM, Greenberg AS, Tjoe KH, Sutherland JS, Chidgey A, van den Brink MRM, Boyd RL. Enhanced immune reconstitution by sex steroid ablation following allogeneic hemopoietic stem cell transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:7473-84. [PMID: 17513799 DOI: 10.4049/jimmunol.178.11.7473] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Delayed immune reconstitution in adult recipients of allogeneic hemopoietic stem cell transplantations (HSCT) is related to age-induced thymic atrophy. Overcoming this paucity of T cell function is a major goal of clinical research but in the context of allogeneic transplants, any strategy must not exacerbate graft-vs-host disease (GVHD) yet ideally retain graft-vs-tumor (GVT) effects. We have shown sex steroid ablation reverses thymic atrophy and enhances T cell recovery in aged animals and in congenic bone marrow (BM) transplant but the latter does not have the complications of allogeneic T cell reactivity. We have examined whether sex steroid ablation promoted hemopoietic and T cell recovery following allogeneic HSCT and whether this benefit was negated by enhanced GVHD. BM and thymic cell numbers were significantly increased at 14 and 28 days after HSCT in castrated mice compared with sham-castrated controls. In the thymus, the numbers of donor-derived thymocytes and dendritic cells were significantly increased after HSCT and castration; donor-derived BM precursors and developing B cells were also significantly increased. Importantly, despite restoring T cell function, sex steroid inhibition did not exacerbate the development of GVHD or ameliorate GVT activity. Finally, IL-7 treatment in combination with castration had an additive effect on thymic cellularity following HSCT. These results indicate that sex steroid ablation can profoundly enhance thymic and hemopoietic recovery following allogeneic HSCT without increasing GVHD and maintaining GVT.
Collapse
Affiliation(s)
- Gabrielle L Goldberg
- Department of Pathology and Immunology, Central and Eastern Clinical School, Monash University, Melbourne, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Jiménez M, Ercilla G, Martínez C. Immune reconstitution after allogeneic stem cell transplantation with reduced-intensity conditioning regimens. Leukemia 2007; 21:1628-37. [PMID: 17525730 DOI: 10.1038/sj.leu.2404681] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Reduced-intensity conditioning (RIC) regimens have been increasingly used as an alternative to conventional myeloablative conditioning (MAC) regimens for elderly patients, for patients medically infirm to qualify for conventional allogeneic stem cell transplantation (SCT), and for disorders in which traditional MAC-SCT are associated with high rates of non-relapse mortality. One of the theoretical advantages of RIC-SCT is that it might lend to better immune reconstitution after transplantation due to less damage of the thymus, allowing regeneration of naive T cells derived from prethymic donor stem cells, and due to the proliferation of immunologically competent host T cells that survive the conditioning regimen. Although limited, studies comparing immune recovery following RIC and MAC-SCT have been insightful. One of the main difficulties of these studies is the current spectrum of RIC protocols, which vary considerably in myeloablative and immunosuppressive potential, resulting in apparently contradictory findings. In spite of this, most reports have shown significant quantitative and/or qualitative differences in T- and B-cell reconstitution after RIC-SCT in comparison with conventional SCT. This paper will review current knowledge of immune reconstitution following RIC-SCT.
Collapse
Affiliation(s)
- M Jiménez
- Department of Hematology, Institute of Hematology and Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer, IDIBAPS, Hospital Clínic, University of Barcelona, Barcelona, Spain.
| | | | | |
Collapse
|
26
|
Ghattas H, Wallace DL, Solon JA, Henson SM, Zhang Y, Ngom PT, Aspinall R, Morgan G, Griffin GE, Prentice AM, Macallan DC. Long-term effects of perinatal nutrition on T lymphocyte kinetics in young Gambian men. Am J Clin Nutr 2007; 85:480-7. [PMID: 17284747 DOI: 10.1093/ajcn/85.2.480] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Nutritional status is highly dependent on season in countries such as The Gambia. In a rural Gambian setting, individuals born during periods of seasonal nutritional deprivation ("hungry seasons") are susceptible to mortality from infectious diseases in adult life. OBJECTIVE We investigated the hypothesis that impaired immunocompetence in those born in the hungry season results from an underlying defect in immunologic memory, similar to the immunosenescence of old age, which is likely to be reflected in the phenotype and kinetics of T lymphocytes in young adults. DESIGN T cell phenotype in terms of CD3, CD4, CD8, CD45RA, and CD45R0 expression and in vivo dynamics measured by stable isotope labeling of T cell subsets combined with gas chromatography-mass spectrometry and frequency of T cell receptor excision circles were measured in 25 young (18-24-y-old) Gambian men. Thirteen of these 25 men were exposed to perinatal malnutrition as defined by birth season and birth weight. RESULTS In persons born in the hungry season with low birth weight, no differences in the proportions of memory or naive T cells were found. Kinetic analysis showed higher proliferation rates in memory (CD45R0(+)) subsets of T cells than in naïve (CD45R0(-)) cells, which is consistent with previous studies, but no evidence was found for an effect of birth weight or season on T lymphocyte proliferation and disappearance rates. No significant correlations were found between in vivo T cell kinetics and frequency of T cell receptor excision circles. Only absolute numbers of granulocytes were elevated in those born in the nutritionally deprived season. CONCLUSION In healthy young Gambian men, T lymphocyte homeostasis is extremely robust regardless of perinatal nutritional compromise.
Collapse
Affiliation(s)
- Hala Ghattas
- Centre for Infection, St George's, University of London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Jiménez M, Martínez C, Ercilla G, Carreras E, Urbano-Ispízua A, Aymerich M, Villamor N, Amézaga N, Rovira M, Fernández-Avilés F, Montserrat E. Clinical factors influencing T-cell receptor excision circle (TRECs) counts following allogeneic stem cell transplantation in adults. Transpl Immunol 2006; 16:52-9. [PMID: 16701177 DOI: 10.1016/j.trim.2006.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Accepted: 02/24/2006] [Indexed: 11/20/2022]
Abstract
To ascertain the clinical factors involved in T-cell reconstitution after allogeneic stem cell transplantation (SCT), we evaluated serial assessments of lymphocyte subsets by flow cytometry and TRECs levels by quantitative PCR in 83 adult patients. Patient age >25 years, unrelated donor, CMV infection and acute graft-versus-host disease (GVHD) adversely affected CD3(+) and CD8(+) T-cell recovery after SCT (p < 0.05). TRECs were low or undetectable during the first months after transplant and progressively increased thereafter. However, median TRECs of patients did never achieve normal values compared to healthy donors (median follow-up 9 months, range 2-42). Presence and severity of chronic GVHD significantly affected TRECs counts: patients with chronic GVHD had lower TRECs than patients without GVHD at 9, 12 and 24 months after SCT (p = 0.002, p = 0.022, p = 0.015). Patients with limited chronic GVHD had higher TRECs compared to patients with extensive GVHD (p = 0.018). No relationship was observed between fungal or bacterial infections and TRECs. Nonetheless, CMV infection was associated with lower TRECs (p = 0.032). Our data support the concept that adult thymus contributes with a slow but continuous production of thymic T cells to immune reconstitution after SCT. Chronic GVHD is the main factor associated to a delay in TRECs counts recovery.
Collapse
MESH Headings
- Adult
- Cells, Cultured
- Female
- Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor
- Gene Rearrangement, delta-Chain T-Cell Antigen Receptor
- Graft vs Host Disease/immunology
- Humans
- Immunophenotyping
- Lymphocyte Count
- Male
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/biosynthesis
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Stem Cell Transplantation/adverse effects
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Transplantation, Homologous
Collapse
Affiliation(s)
- Mónica Jiménez
- Department of Hematology, Institute of Hematology and Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer, IDIBAPS, Hospital Clínic, University of Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Jelley-Gibbs DM, Brown DM, Dibble JP, Haynes L, Eaton SM, Swain SL. Unexpected prolonged presentation of influenza antigens promotes CD4 T cell memory generation. ACTA ACUST UNITED AC 2006; 202:697-706. [PMID: 16147980 PMCID: PMC2212871 DOI: 10.1084/jem.20050227] [Citation(s) in RCA: 211] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kinetics of presentation of influenza virus–derived antigens (Ags), resulting in CD4 T cell effector and memory generation, remains undefined. Naive influenza-specific CD4 T cells were transferred into mice at various times after influenza infection to determine the duration and impact of virus-derived Ag presentation. Ag-specific T cell responses were generated even when the donor T cells were transferred 3–4 wk after viral clearance. Transfer of naive CD4 T cells during early phases of infection resulted in a robust expansion of highly differentiated effectors, which then contracted to a small number of memory T cells. Importantly, T cell transfer during later phases of infection resulted in a modest expansion of effectors with intermediate phenotypes, which were capable of persisting as memory with high efficiency. Thus, distinct stages of pathogen-derived Ag presentation may provide a mechanism by which T cell heterogeneity is generated and diverse memory subsets are maintained.
Collapse
|
29
|
Abstract
Thymic function is important for the generation of T-cell diversity in the periphery of both children and adults during both health and disease. Until recently, thymic function could not be monitored, as a consequence of the absence of adequate technology to differentiate recent thymic emigrants (RTEs) from naïve T cells. The generation of TCR diversity occurs in the thymus through recombination of gene segments encoding the variable parts of the TCR alpha and beta chains. During these processes, by-products of the rearrangements are generated in the form of signal joint T-cell receptor excision circles (sjTRECs). As sjTRECs are stable extrachromosomal DNA fragments, they are not replicated during mitosis and thus diluted with each round of cell division, and are therefore most frequent in naïve T cells that have recently left the thymus, their quantification is actually considered as a valuable tool to estimate thymic function. Therefore, quantitative sjTRECs content have been recently used to assess thymic output during both health and disease. In this review, we summarize recent data on the recent thymic output function feature in patients with hematological malignancy and the immune reconstitution after stem cell transplantation and we also characterize factors that may improve the thymic output function.
Collapse
Affiliation(s)
- Yangqiu Li
- Institute of Hematology, Medical College, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
30
|
Goldberg GL, Sutherland JS, Hammet MV, Milton MK, Heng TSP, Chidgey AP, Boyd RL. Sex steroid ablation enhances lymphoid recovery following autologous hematopoietic stem cell transplantation. Transplantation 2005; 80:1604-13. [PMID: 16371932 DOI: 10.1097/01.tp.0000183962.64777.da] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Autologous hematopoietic stem cell transplantation (auto-HSCT) patients experience long-term immunosuppression, which increases susceptibility to infection and relapse rates due to minimal residual disease (MRD). Sex steroid (SS) ablation is known to reverse age-related thymic atrophy and decline in B-cell production METHODS This study used a congenic HSCT mouse model to analyze the effects of SS ablation (through surgical castration) on immune reconstitution and growth factor production following auto-HSCT. Bone marrow (BM) and thymic stromal cell (TSCs) populations were analyzed using RT-PCR and were tested for the production of growth factors previously implicated in immune reconstitution or age-relate immune degeneration RESULTS Castration increased bone marrow (BM), thymic, and splenic cellularity following auto-HSCT. HSC number and common lymphoid precursor (CLP) frequency and number were increased in castrated mice. B cell precursor numbers were also significantly increased in the BM of these mice. Triple negative, double positive and single positive thymocytes were increased following HSCT and castration, as were thymic dendritic cells and natural killer T (NKT) cells. This enhanced lymphoid reconstitution of the primary immune organs leads to a significant increase in splenic T and B cells 42 days after HSCT. The molecular mechanisms behind the enhanced reconstitution were also studied. TGF-beta1 was decreased in castrated mice compared to sham-castrated controls in TSCs and BM cells. TSC production of IL-6 was also decreased in castrated mice CONCLUSIONS These data suggest that sex steroid ablation significantly enhances lymphopoiesis following auto-HSCT providing a new strategy for posttransplant immune reconstitution.
Collapse
Affiliation(s)
- Gabrielle L Goldberg
- Department of Immunology, Central and Eastern Clinical School, Monash University, Melbourne, Australia
| | | | | | | | | | | | | |
Collapse
|
31
|
Jiménez M, Martínez C, Ercilla G, Carreras E, Urbano-Ispízua A, Aymerich M, Villamor N, Amézaga N, Rovira M, Fernández-Avilés F, Gaya A, Martino R, Sierra J, Montserrat E. Reduced-intensity conditioning regimen preserves thymic function in the early period after hematopoietic stem cell transplantation. Exp Hematol 2005; 33:1240-8. [PMID: 16219547 DOI: 10.1016/j.exphem.2005.06.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2005] [Revised: 06/07/2005] [Accepted: 06/08/2005] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To compare T-cell reconstitution in two groups of patients submitted to allogeneic stem cell transplantation (SCT): those receiving reduced-intensity conditioning (RIC, n = 24) and those receiving myeloablative conditioning (MA, n = 27). METHODS Fifty-one consecutive patients undergoing SCT were evaluated. Serial assessments of lymphocyte subsets and T cell receptor excision circles (TRECs) levels were performed using multiparametric flow cytometry and real-time PCR, respectively. RESULTS During the first 6 months posttransplant, total and naïve CD4(+) T cell counts were higher after RIC-SCT than after MA-SCT (total CD4(+): p = 0.04, p = 0.08, and p = 0.058; naïve CD4(+): p = 0.14, p = 0.05, and p = 0.01 at 1, 3, and 6 months, respectively). In both groups of patients, TRECs levels were low or undetectable in the first 3 months after SCT and progressively increased during the study. However, a higher proportion of patients with detectable levels of TRECs was observed in RIC-SCT at 1 and 3 months and more patients in this group reached normal levels of TRECs at 6 months post-SCT. In the multivariate analysis, including factors such as type of donor (sibling vs unrelated), dose of CD34(+) cells infused with the graft, patient age, and graft-vs-host disease (GVHD), the most important factor influencing TRECs recovery in the early period after SCT was the type of conditioning regimen. CONCLUSIONS In this study, the pattern of immune reconstitution after RIC-SCT was different from that of MA-SCT and was characterized by higher posttransplant naïve CD4(+) T cell counts and TRECs levels in the early period after transplant.
Collapse
Affiliation(s)
- Mónica Jiménez
- Department of Hematology, Institute of Hematology and Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer, IDIBAPS, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Clave E, Rocha V, Talvensaari K, Busson M, Douay C, Appert ML, Rabian C, Carmagnat M, Garnier F, Filion A, Socié G, Gluckman E, Charron D, Toubert A. Prognostic value of pretransplantation host thymic function in HLA-identical sibling hematopoietic stem cell transplantation. Blood 2005; 105:2608-13. [PMID: 15546951 DOI: 10.1182/blood-2004-04-1667] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Abstract
Thymic function is critical for immune reconstitution after hematopoietic stem cell transplantation (HSCT). We evaluated recipient thymic function before HSCT by quantifying T-cell receptor excision circles (TRECs) in pretransplantation peripheral blood lymphocytes from 102 patients who received HSCs from an HLA-identical sibling for malignant (n = 87) or nonmalignant diseases (n = 15). Median TREC value before transplantation was 257 TRECs per 150 000 CD3+ cells (range, 0-42 746). We assessed 172 TRECs per 150 000 CD3+ cells as the most discriminating TREC value for survival in a first cohort of patients (n = 62). This cut-off was validated in a second independent prospective group of 40 patients. In the 102 patients, a TREC value greater than or equal to 172 was associated with a better survival (P < .000 01), a decreased incidence of grade II-IV acute graft-versus-host disease (GVHD; P = .017), chronic GVHD (P = .023), and bacterial (P = .003) and cytomegalovirus (CMV) infection (P = .024). In a multivariate analysis, low pretransplantation TREC values were associated with a higher incidence of CMV infection (hazard ratio [HR] = 2.0, P = .06) and severe bacterial infections (HR = 2.8, P = .036). Finally, high TREC values (HR = 6.6, P = .002) and ABO compatibility (HR = 2.7, P = .02) were associated with a better survival. Therefore, recipient host thymic function assessment could be helpful in predicting HSCT outcome and identifying patients who require a close immunologic monitoring.
Collapse
Affiliation(s)
- Emmanuel Clave
- Laboratoire d'Immunologie et d'Histocompatibilité Assistance Publique-Hôpitaux de Paris (AP-HP), Institut National de la Santé et de la Recherche Médicale (INSERM) U396, Hôpital Saint-Louis, Paris, CEDEX 10, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen X, Barfield R, Benaim E, Leung W, Knowles J, Lawrence D, Otto M, Shurtleff SA, Neale GAM, Behm FG, Turner V, Handgretinger R. Prediction of T-cell reconstitution by assessment of T-cell receptor excision circle before allogeneic hematopoietic stem cell transplantation in pediatric patients. Blood 2005; 105:886-93. [PMID: 15358630 DOI: 10.1182/blood-2004-04-1405] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
The extent and rapidity with which T cells are regenerated from graft-derived precursor cells directly influences the incidence of infection and the T-cell–based graft-versus-tumor effect. Measurement of T-cell receptor excision circles (TRECs) in peripheral blood is a means of quantifying recent thymic T-cell production and has been used after transplantation in many studies to estimate thymus-dependent T-cell reconstitution. We hypothesized that the quality of thymic function before transplantation affects thymus-dependent T-cell reconstitution after transplantation. We used real-time polymerase chain reaction (PCR) to quantify signal-joint TRECs (sjTRECs) before and after transplantation. T-cell reconstitution was evaluated by T-cell receptor β (TCRβ) CDR3 size spectratyping. We tested 77 healthy sibling donors and 244 samples from 26 pediatric recipients of allogeneic hematopoietic stem cell transplantation (AHSCT). Blood from the healthy donors contained 1200 to 155 000 sjTREC copies/mL blood. Patients who had greater than 1200 copies/mL blood before transplantation showed early recovery of sjTREC numbers and TCRβ repertoire diversity. In contrast, patients who had fewer than 1200 copies/mL blood before transplantation demonstrated significantly slower restoration of thymus-dependent T cells. We conclude that the rate of reconstitution of thymus-dependent T cells is dependent on the competence of thymic function in the recipients before transplantation. Therefore, pretransplantation measurement of sjTREC may provide an important tool for predicting thymus-dependent T-cell reconstitution after transplantation.
Collapse
Affiliation(s)
- Xiaohua Chen
- Division of Stem Cell Transplantation, St Jude Children's Research Hospital, Memphis TN 38105-2794, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Halnon NJ, Jamieson B, Plunkett M, Kitchen CMR, Pham T, Krogstad P. Thymic function and impaired maintenance of peripheral T cell populations in children with congenital heart disease and surgical thymectomy. Pediatr Res 2005; 57:42-8. [PMID: 15531736 DOI: 10.1203/01.pdr.0000147735.19342.de] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The thymus begins involution in childhood and historically it was thought to be nonfunctional by adulthood, thus presenting no contraindication to the routine practice of thymectomy during cardiothoracic surgery. More recent data suggest, however, that the thymus remains active into adulthood and is responsible for the low-level production of normal T cells. We hypothesize, therefore, that incidental thymectomy during cardiothoracic surgery in infancy causes long-term changes in the cellular immune system. To investigate this hypothesis, we quantified peripheral T-cell subsets and T-cell recombination excision circles in children with congenital heart disease to measure the impact of cardiothoracic surgical procedures and thymectomy performed during a period of immunologic development. We found that cardiothoracic surgical procedures, especially if they include thymectomy, impair T-cell production and produce long-term decreases in total lymphocyte count and CD4(+) and CD8(+) T-cell subsets, suggesting that long-term maintenance of lymphocyte populations is disturbed.
Collapse
Affiliation(s)
- Nancy J Halnon
- Division of Pediatric, David Geffen School of Medicine at the University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|
35
|
Bordi L, Amendola A, Ciccosanti F, Abbate I, Camilloni G, Capobianchi MR. Expression of Werner and Bloom syndrome genes is differentially regulated by in vitro HIV-1 infection of peripheral blood mononuclear cells. Clin Exp Immunol 2004; 138:251-8. [PMID: 15498034 PMCID: PMC1809202 DOI: 10.1111/j.1365-2249.2004.02622.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In HIV infection, continuous immune activation leads to accelerated ageing of the adaptive immune system, similar to that observed in elderly people. We investigated the expression of WRN and BLM (genes involved in disorders characterized by premature ageing, genomic instability and cancer predisposition) in peripheral blood mononuclear cells (PBMC) activated in vitro with phytohaemagglutinin (PHA) and infected with different HIV-1 strains. The steady state levels of mRNA were analysed by reverse transcription-polymerase chain reaction (RT-PCR), and protein expression was assayed using immunocytochemistry and Western blot techniques. In uninfected PBMC, PHA stimulation induced an increase in BLM mRNA and protein expression, while WRN expression remained virtually unchanged. When PBMC were infected in vitro with a lymphotropic HIV-1 strain, the level of BLM mRNA showed a peak at 24 h of infection, followed by a decline to uninfected culture levels. A similar result failed to be seen using an R5-tropic HIV-1 strain. In accordance with mRNA expression, in HIV-infected cultures PBMC were stained more frequently and more intensely by a BLM-specific antibody as compared to uninfected cultures, staining peaking at 24. Conversely, WRN expression was not modulated by HIV-1. The proportion of cells showing BLM up-regulation, established by immunocytochemical staining, was much greater than the proportion of productively infected PBMC, as established by proviral DNA measurement. This result indicates that BLM up-regulation is probably a result of an indirect bystander cell effect. Activation of the BLM gene in infected PBMC suggests that premature ageing could be a further immunopathogenetic mechanism involved in HIV-induced immunodeficiency, and points to a possible new candidate target for innovative therapeutic intervention.
Collapse
Affiliation(s)
- L Bordi
- National Institute for Infectious Diseases 'L. Spallanzani', Università di Roma 'La Sapienza', Istitito di Biologia e Patologia Molecolare CNR, Rome, Italy
| | | | | | | | | | | |
Collapse
|
36
|
van den Brink MRM, Alpdogan O, Boyd RL. Strategies to enhance T-cell reconstitution in immunocompromised patients. Nat Rev Immunol 2004; 4:856-67. [PMID: 15516965 DOI: 10.1038/nri1484] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Immune deficiency, together with its associated risks such as infections, is becoming an increasingly important clinical problem owing to the ageing of the general population and the increasing number of patients with HIV/AIDS, malignancies (especially those treated with intensive chemotherapy or radiotherapy) or transplants (of either solid organs or haematopoietic stem cells). Of all immune cells, T cells are the most often affected, leading to a prolonged deficiency of T cells, which has important clinical consequences. Accordingly, strategies to improve the recovery and function of T cells, as we discuss here, should have a direct impact on reducing the morbidity and mortality of many patients and should increase the efficacy of therapeutic and prophylactic vaccinations against microbial pathogens or tumours.
Collapse
Affiliation(s)
- Marcel R M van den Brink
- Departments of Medicine and Immunology, Box 111-Kettering 406D, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA.
| | | | | |
Collapse
|
37
|
De Vera MJ, Al-Harthi L, Gewurz AT. Assessing thymopoiesis in patients with common variable immunodeficiency as measured by T-cell receptor excision circles. Ann Allergy Asthma Immunol 2004; 93:478-84. [PMID: 15562888 DOI: 10.1016/s1081-1206(10)61416-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Common variable immunodeficiency (CVID) is one of the most common primary immune deficiencies. The immunologic hallmark of CVID is failure of B-cell differentiation and impaired secretion of immunoglobulins. However, there is mounting evidence of accompanying T-cell dysregulation, which could be due to abnormal thymic function because the thymus plays a crucial role in T-cell development. Recently, it was shown that the human thymus remains functional well into adulthood. Current data show that the level of T-cell receptor excision circles (TRECs) correlates well with active thymopoiesis. OBJECTIVE To determine whether thymic dysfunction contributes to the pathogenesis of CVID. METHODS We evaluated 15 patients, aged 19 to 65 years, previously diagnosed as having CVID. Genomic DNA was isolated from peripheral blood mononuclear cells of each patient. Thymic output was evaluated by measuring coding joint TRECs in the total T-cell population using real-time polymerase chain reaction. RESULTS Results were compared with known age-matched reference values. The median TREC level in patients with CVID (82,034 copies/microg of DNA) was significantly higher than that in the healthy cohort (43,000 copies/microg of DNA) (P < .001). In examining the relationship between TREC levels and age, we noted that TREC levels significantly declined faster with age in patients with CVID vs the healthy cohort. CONCLUSIONS In these patients, thymic dysregulation may be a factor in CVID, with an accelerated rate of TREC loss with age compared with healthy adults.
Collapse
Affiliation(s)
- Michelle J De Vera
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | |
Collapse
|
38
|
Baev DV, Peng XH, Song L, Barnhart JR, Crooks GM, Weinberg KI, Metelitsa LS. Distinct homeostatic requirements of CD4+ and CD4- subsets of Valpha24-invariant natural killer T cells in humans. Blood 2004; 104:4150-6. [PMID: 15328159 DOI: 10.1182/blood-2004-04-1629] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
CD1d-restricted Valpha24-invariant natural killer T cells (iNKTs) are important in immunoregulation. CD4(+) and CD4(-) iNKTs develop with similar frequencies in murine thymus and depend on interleukin-15 (IL-15) in periphery. However, homeostatic requirements of iNKTs have not been analyzed in humans. We evaluated thymic production, peripheral dynamics, and functional maturation of human iNKTs. CD4(+) subset comprises 90% of iNKTs in mature thymocytes and cord blood (CB) but only 40% in adult blood. Using T-cell receptor excision circle (TREC) analysis, we directly measured in vivo replicative history of CD4(+) and CD4(-) iNKT cells. Compared to CD4(+), CD4(-) iNKTs contain fewer TRECs, express higher levels of IL-2Rbeta, and proliferate with higher rate in response to IL-15. In contrast, CD4(+) cells express higher levels of IL-7Ralpha and better respond to IL-7. Neither thymic nor CB iNKTs are able to produce cytokines unless they are induced to proliferate. Therefore, unlike in the mouse, human CD4(+) iNKTs are mainly supported by thymic output and limited peripheral expansion, whereas CD4(-) cells undergo extensive peripheral expansion, and both subsets develop their functions in periphery. These findings reveal important differences in homeostatic requirements and functional maturation between murine and human iNKTs that are to be considered for clinical purposes.
Collapse
Affiliation(s)
- Denis V Baev
- Division of Hematology-Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, Keck School of Medicine, University of Southern California, SRT-501-d, 4650 Sunset Boulevard, Los Angeles, CA 90027, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Al-Harthi L, Voris J, Patterson BK, Becker S, Eron J, Smith KY, D'Amico R, Mildvan D, Snidow J, Pobiner B, Yau L, Landay A. Evaluation of the impact of highly active antiretroviral therapy on immune recovery in antiretroviral naive patients. HIV Med 2004; 5:55-65. [PMID: 14731171 DOI: 10.1111/j.1468-1293.2004.00186.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To examine the extent of immune reconstitution in treatment-naive patients with CD4 T-cell counts <500 cells/microL following 48 weeks of highly active antiretroviral therapy (HAART). METHODS Thirteen antiretroviral naive patients were evaluated longitudinally for 48 weeks on HAART utilizing immune functional and lymphocyte phenotyping assays, including lymphocyte proliferation assay, flow cytometric evaluation of cell surface markers, and delayed type hypersensitivity skin tests. Virologic responses were monitored using commercially available viral load assays and gag/pol mRNA quantification using simultaneous immunophenotyping/UltraSensitive fluorescence in situ hybridization (ViroTect In Cell HIV-1 Detection Kit; Invirion, Frankfort, MI). Thymic function was evaluated for a subset of four patients using real-time polymerase chain reaction (PCR) for T-cell receptor excision circle (TREC) quantification and thymic scans using computerized axial tomography (CT) of the thymus. RESULTS HAART initiation resulted in a significant decline in plasma viremia and percentage of infected peripheral blood cells, and a rise in CD4 T cells from a baseline median of 207 cells/microL to a week-48 median of 617 cells/microL. The rise was predominately in CD4 memory cells. Naive T cells also increased in number, but at a slower rate. Activated (HLA-DR CD38) CD4 and CD8 T cells were elevated at baseline (24 and 62%, respectively) and declined by week 48 (17 and 36%, respectively) but did not reach normal levels. The number of Fas CD4 T cells increased from a baseline median of 169 to 381 cells/microL at week 48. Both soluble interleukin (IL)-2 and tumour necrosis factor (TNF) II receptors declined by week 48. HIV p24 lymphocyte proliferation assay responses were transiently detected in three patients. TREC values increased from a median 6400 copies/microg at baseline to a week-48 median value of 26 697 copies/microg. CONCLUSION Immune functional reconstitution was not achieved in these HAART naive patients.
Collapse
Affiliation(s)
- L Al-Harthi
- Department of Immunology/Microbiology, Rush-Presbyterian- St. Luke's Medical Center, Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Richardson MW, Sverstiuk AE, Silvera P, Greenhouse J, Lisziewicz J, Lori F, Khalili K, Lewis MG, Rappaport J. T-cell receptor excision circles (TREC) in SHIV 89.6p and SIVmac251 models of HIV-1 infection. DNA Cell Biol 2004; 23:1-13. [PMID: 14965468 DOI: 10.1089/104454904322745880] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
T-cell receptor excision circles (TREC) may be a useful surrogate marker in HIV-1 infection for evaluating the likelihood of continued clinical stability and/or the response to therapeutics, including vaccines. Analysis of TREC in SHIV and SIV models of HIV-1 infection may provide additional information concerning the utility of TREC as a marker. We measured TREC in peripheral blood mononuclear cells (PBMC) from rhesus macaques in SHIV89.6p (n = 20) and SIVmac251 (n = 11) models of HIV-1 infection. TREC were also evaluated in tissues in the SIVmac251 model at end-point. In the SHIV89.6p model, TREC in PBMC were significantly lower at 12 weeks postinfection compared to preinfection levels. The decrease in TREC correlated with the decline in CD4+ T cells (r(s) = 0.496; P = 0.026), which in turn correlated inversely with serum viral loads at end-point (r(s) = -0.517; P = 0.019). Macaques that controlled SHIV89.6p infection to some degree (n = 6) had higher TREC at study end-point (P = 0.017). In the SIVmac251 model, TREC in PBMC were significantly reduced after 17 months of infection (P = 0.012) despite receiving highly active antiretroviral therapy (HAART) consisting of didanosine (ddI) and (R)-9-(2-phosphonylmethoxypropyl)-adenine (PMPA) when not cycling off therapy during scheduled treatment interruptions (STI). However, macaques that received continuous hydroxyurea (HU) in addition to the HAART regimen had higher end-point TREC compared to the non-HU group (P = 0.041), and the reduction in TREC observed at end-point within the HU group was not significant. In the SIVmac251 model, TREC correlated with the percentage of CD4+ T cells (r(s) = 0.426; P = 0.048) and CD4+CD28+ T cells (r(s) = 0.624; P = 0.002), and inversely with CD8+ T cells (r(s) = -0.622; P = 0.002), CD8+CD28- T cells (r(s) = -0.516; P = 0.014), and serum viral loads (r(s) = -0.627; P = 0.039). High levels of TREC were observed in the thymus, levels comparable to PBMC were seen in the lymph node, and low but detectable levels of TREC were present in bone marrow. The use of correlates of TREC as covariates in ANCOVA revealed that the decline in TREC in the SHIV 89.6p model reflected the decline in the percentage of CD4+ T-cells due to viral cytopathogenicity. In the SIVmac251 model, the decline in TREC was related to increased immune activation and proliferation due to viral replication, as reflected by decreases in percentages of CD4+CD28+ T cells and increases in CD8+ and CD8+CD28- T cells.
Collapse
Affiliation(s)
- Max W Richardson
- Center for Neurovirology and Cancer Biology, Temple University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Henson SM, Pido-Lopez J, Aspinall R. Reversal of thymic atrophy. Exp Gerontol 2004; 39:673-8. [PMID: 15050305 DOI: 10.1016/j.exger.2003.10.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2003] [Revised: 09/25/2003] [Accepted: 10/10/2003] [Indexed: 11/15/2022]
Abstract
Age-associated thymic atrophy is a key event preceding the inefficient functioning of the immune system, resulting in a diminished capacity to generate new T-cells. This thymic involution has been proposed to be due to changes in the thymic microenvironment resulting in its failure to support thymopoiesis. A key cytokine in the early stages of thymocyte development is IL-7 and expression levels are greatly reduced with age. The ability of IL-7 to restore the immune system by enhancing thymic output remains controversial. In this review, we highlight the advances in molecular approaches used to evaluate recent thymic emigrants and assess the success of these strategies in determining whether IL-7 can lead to immune reconstitution.
Collapse
Affiliation(s)
- Sian M Henson
- Department of Immunology, Imperial College, Chelsea and Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK.
| | | | | |
Collapse
|
42
|
Wheeler CJ, Black KL, Liu G, Ying H, Yu JS, Zhang W, Lee PK. Thymic CD8+ T cell production strongly influences tumor antigen recognition and age-dependent glioma mortality. THE JOURNAL OF IMMUNOLOGY 2004; 171:4927-33. [PMID: 14568975 DOI: 10.4049/jimmunol.171.9.4927] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
For unknown reasons, advanced age remains a dominant predictor of poor clinical outcome for nearly all cancers. A decrease in the production of T cells by the thymus accompanies normal aging and parallels the age-dependent increase in cancer progression, but the specific impact of immunity on tumor progression in general is unknown. Glioblastoma multiforme (GBM), the most common primary brain neoplasm, is characterized by rapid age-dependent rates of progression and death. In this study, we show levels of CD8(+) recent thymic emigrants (RTEs) accounted for the prognostic power of age on clinical outcome in GBM patients. CD8(+) RTEs, typically a tiny proportion of CD8(+) T cells, remarkably accounted for the majority of tumor Ag-binding small precursor cells in PBMC from these patients and from healthy individuals. Large blasting tumor Ag-binding cells comprised of CD8(+) RTEs and phenotypically related cells were predominantly expanded following experimental vaccination of GBM patients. Quantification of CD8(+) RTE expansion in vivo correlated strongly with vaccine-elicited cytokine responses, and estimated numbers of expanding CD8(+) RTEs were consistent predictors of clinical outcome in vaccinated GBM patients. Targeted mutant (CD8beta(-/-)) mice specifically deficient in thymic CD8(+) T cell production uniquely displayed an age-specific decrease in glioma host survival as well as a strong correlation between host survival and thymus cellular production. These findings suggest that levels and function of newly produced CD8(+) T cells critically influence age-dependent cancer mortality and exert one of the strongest known influences on GBM outcome by predominantly mediating clinically beneficial antitumor immune responses.
Collapse
Affiliation(s)
- Christopher J Wheeler
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Murray JM, Kaufmann GR, Hodgkin PD, Lewin SR, Kelleher AD, Davenport MP, Zaunders JJ. Naive T cells are maintained by thymic output in early ages but by proliferation without phenotypic change after age twenty. Immunol Cell Biol 2003; 81:487-95. [PMID: 14636246 DOI: 10.1046/j.1440-1711.2003.01191.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Analysing T-cell receptor excision circle numbers in healthy individuals we find a marked change in the source of naive T cells before and after 20 years of age. The bulk of the naive T cell pool is sustained primarily from thymic output for individuals younger than 20 years of age whereas proliferation within the naive phenotype is dominant for older individuals. Over 90% of phenotypically naive T cells in middle age are not of direct thymic origin. Moreover, this change in source of naive T cells is accompanied either by an increased death rate of T cells from the thymus or reduced thymic export. Modelling of these processes shows that new naive T cells of a thymic origin have a half-life of approximately 50 days before this change occurs, and that either the life-span of recent thymic emigrants (but not necessarily of all naive cells) decreases approximately threefold in middle age, or thymic production drops by this same amount. The decay rate of T-cell receptor excision circle levels for individuals over 20 years of age is consistent with the decay rate of the productive thymus. Our modelling suggests that at age 25, thymic export is responsible for 20% of naive T-cell production and that this percentage decreases with the 15.7 year half-life of the productive thymus so that by age 55 only 5% of naive production arises from thymic export.
Collapse
Affiliation(s)
- John M Murray
- School of Mathematics, University of NSW, Sydney, Australia.
| | | | | | | | | | | | | |
Collapse
|
44
|
Domínguez-Gerpe L, Rey-Méndez M. Evolution of the thymus size in response to physiological and random events throughout life. Microsc Res Tech 2003; 62:464-76. [PMID: 14635139 DOI: 10.1002/jemt.10408] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During embryogenesis and in the early stages of life, the thymus is a crucial organ for the generation of the T cell repertoire. T cells are generated from hematopoietic stem cells already differentiated to precursor T cells in the bone marrow. These cells enter the thymus guided by chemotactic factors secreted by this organ. The complex maturation process takes place that ensures self-tolerance and homeostasis. Thymocytes that show autoreactivity do not leave the thymus, but rather die by apoptosis. The final percentage of mature T cells that survive to migrate from the thymus to the periphery is very low: at most 5%, under optimal conditions. The highest migration occurs in childhood and adulthood, at least in mice and humans; however, it declines throughout life and is minimal in the elderly. Under normal circumstances, the thymus commences involution soon after birth, and this involution correlates with the capacity to export mature T cells to the periphery. Hormones, cytokines, and neurotransmitters all play a role in this age-associated process, but the reasons for and mechanisms of this involution remain unknown. Apart from physiological conditions that change throughout life and govern age-related thymus evolution, random states and events provoked by intrinsic or extrinsic factors can induce either thymus involution, as in reversible transient thymic hypoplasias, or thymic hyperplasias. The age-associated involution, unlike transient involutions, follows a regular pattern for all individuals, though there are clear differences between the sexes. Nevertheless, even the age-associated involution seems to be reversible, raising the possibility of therapeutic strategies aimed at enhancing thymus function in the elderly.
Collapse
Affiliation(s)
- Lourdes Domínguez-Gerpe
- Departamento de Bioquímica y Biología Molecular, Facultad de Biología, Universidad de Santiago de Compostela, 15706 Santiago de Compostela, La Coruña, Spain.
| | | |
Collapse
|
45
|
Svaldi M, Lanthaler AJ, Dugas M, Lohse P, Pescosta N, Straka C, Mitterer M. T-cell receptor excision circles: a novel prognostic parameter for the outcome of transplantation in multiple myeloma patients. Br J Haematol 2003; 122:795-801. [PMID: 12930391 DOI: 10.1046/j.1365-2141.2003.04482.x] [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] [Indexed: 11/20/2022]
Abstract
This study investigated whether T-cell receptor excision circles (TRECs) are a prognostic marker for the outcome of myeloma patients undergoing a tandem autologous peripheral blood stem cell transplantation (PBSCT). Twenty-five patients were enrolled. Samples were obtained at study enrollment, after conventional therapy, between first and second transplantation and 3, 6, 12 and 24 months after the second PBSCT. TRECs were quantified using real-time polymerase chain reaction. A high variation in TREC levels was found at diagnosis (median TREC level 136/10(5) peripheral blood mononuclear cells (PBMCs); range 1-1729), suggesting individual differences in thymic output of naive T cells. Patients with more than 136 TRECs/10(5) P BMCs at diagnosis had a statistically significant better overall survival (P=0.05) and event-free survival (P=0.045), whereas low TREC levels correlated with a higher incidence of infectious complications. Median TREC values were lowest after the first PBSCT (52/10(5) PBMCs) and reached the baseline 12 months after the second transplantation. Patients with high TREC levels after the second PBSCT had a significantly higher probability of being in complete or partial remission 30 months after the second PBSCT. TREC levels were not correlated with beta2-microglobulin and C-reactive protein levels at diagnosis. These data suggest that TRECs could be a relevant prognostic factor for patients who receive high-dose chemotherapy and autologous PBSCT.
Collapse
Affiliation(s)
- Mirija Svaldi
- Department of Hematology and Bone Marrow Transplantation Centre, Regional Hospital, Bozen, Italy.
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Cellular immunodeficiency is associated with human cancer. Extensive reviews on cancer of the head and neck, lung, esophagus and breast convince the author that for these diseases the immunodeficiency is reasonably well established yet the mechanisms are poorly understood. Evidence indicates that other tumors are similarly associated with cellular immune deficiency. The advent of recombinant cytokines and of antitumor monoclonal antibodies has served to focus attention toward direct tumoricidal mechanisms. As tumor antigens relating to cellular and humoral immune mechanisms are being defined and vaccine strategies are increasingly being attempted, it is critical to confront issues of the mechanism of anergy and effective immunorestoration in order to maximize the potential of cellular immune response to address these tumor antigens. Intrinsic to this approach is the introduction of contrasuppressive therapy to alleviate the tumor-associated immune suppression. Encouraging attempts have been made with plasmapheresis, indomethacin, low-dose cyclophosphamide, anti CTLA-4, anti FAS ligand and, perhaps in the future, more judiciously applied chemotherapy. In contrast to the popular notion that thymic involution cannot be reversed in the adult, studies from the author's laboratory indicate that in aged hydrocortisone stressed mice, a natural Type 1-cytokine mixture (IRX-2) hastens the reversal of thymic involution and promotes T-cell responses to cytokines and mitogens. Recombinant IL-1 and IL-2 by themselves, and in combination, were inactive. Similar positive effects were observed with oral zinc, zinc-thymulin and thymosin alpha(1). The combination of a natural cytokine mixture (IRX-2) with thymosin alpha1 had a very large effect and increased the absolute number of peripheral T lymphocytes as measured in the spleen. In studies of combination immunotherapy in lymphocytopenic squamous cell head and neck cancer patients using IRX-2 (18 patients) and IRX-2 plus thymosin alpha(1) (IRX-3) in IRX-2-refractory patients (7 patients), marked increases in CD(45)RA(+) 'naïve' T cells (>250/mm(3)) were observed. These are among the first insights into how to generate T lymphocyte replacement in the adult. These and many other experimental efforts point to ways to achieve more effective immunotherapy of human cancer in the future, particularly if tumor-induced immune deficiency can be effectively addressed.
Collapse
Affiliation(s)
- John W Hadden
- Immuno-Rx, Inc., 140 West 57th Street, Suite 9C, New York, NY 10019, USA
| |
Collapse
|
47
|
Richardson MW, Sverstiuk AE, Gracely EJ, Hendel H, Khalili K, Zagury JF, Rappaport J. T-cell receptor excision circles (TREC) and maintenance of long-term non-progression status in HIV-1 infection. AIDS 2003; 17:915-7. [PMID: 12660540 DOI: 10.1097/00002030-200304110-00018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Max W Richardson
- Center for Neurovirology and Cancer Biology, Temple University, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Pham T, Belzer M, Church JA, Kitchen C, Wilson CM, Douglas SD, Geng Y, Silva M, Mitchell RM, Krogstad P. Assessment of thymic activity in human immunodeficiency virus-negative and -positive adolescents by real-time PCR quantitation of T-cell receptor rearrangement excision circles. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2003; 10:323-8. [PMID: 12626462 PMCID: PMC150534 DOI: 10.1128/cdli.10.2.323-328.2003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Circular DNA molecules known as T-cell receptor rearrangement excision circles (TREC) arise during T-cell development and are present in cells that have recently emigrated from the thymus. In cross-sectional studies, the number of peripheral blood lymphocytes bearing TREC decreases with age, consistent with an anatomically demonstrated loss of thymic epithelial tissue. TREC numbers increase following hematopoietic stem cell transplantation and during therapy for human immunodeficiency virus (HIV) infection. Quantitation of TREC has therefore been proposed as a parameter of thymic activity. In this study, we used real-time PCR to quantify TREC in peripheral blood samples obtained longitudinally from HIV-seronegative adolescents. TREC values in peripheral blood T cells were very stable throughout adolescence, once thought to be a time of rapid involution of the thymus. In addition, in a cross-sectional analysis, we examined TREC values in a cohort of HIV-positive adolescents and found evidence of ongoing thymopoiesis in perinatally infected individuals, despite lifelong infection. These data demonstrate the utility of TREC assessment in adolescents and that HIV infection does not uniformly result in accelerated thymic involution in childhood.
Collapse
Affiliation(s)
- Thao Pham
- David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Nokta MA, Li XD, Al-Harthi L, Nichols J, Pou A, Asmuth D, Landay A, Pollard RB. Entrapment of recent thymic emigrants in lymphoid tissues from HIV-infected patients: association with HIV cellular viral load. AIDS 2002; 16:2119-27. [PMID: 12409732 PMCID: PMC3738871 DOI: 10.1097/00002030-200211080-00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE(S) Depletion of thymus derived naive T-cells is a feature of HIV infection. Here the impact of HIV infection on the compartmentalization of recent thymic emigrants of (RTE) and naive T-cells was examined. METHODS Peripheral blood mononuclear cells (PBMC) and lymphoid tissue (LT) from 43 HIV-infected patients and 12 controls were examined for RTE distribution by measuring coding joint T-cell receptor excisional circles (cjTREC) by PCR and naive and memory T-cell subsets and adhesion molecules (L-selection, LFA-1) by flow cytometry. RESULTS In HIV-infected patients, the RTE as quantified by cjTRECs in CD4 LT cells were significantly higher than in PBMC. Their values, however, were less than in control subjects, in both the LT and PBMC compartments. This was associated with an increase in L-selectin and LFA-1 expression on LT derived T cells. In PBMC, a significant positive relationship between TREC and naive CD4 cells and an inverse relationship between TREC and cellular viral load (CVL) was observed. Whereas in LT, there was a positive relationship between cjTREC and both naive CD4 cell percentage and CVL. CONCLUSIONS Collectively, the data suggests that LT is a significant reservoir for RTE. The RTE appeared to be entrapped in LT from HIV-infected subjects. Such entrapment is probably a response to the high viral load in these tissues. These observations may partially explain the decline in RTE observed in the peripheral blood of HIV-infected patients, and the delay in recovery of naive cells in blood after initiation of HAART.
Collapse
|
50
|
Abstract
The reconstitution of T-cell populations is a critical component of immune recovery after allogeneic stem cell transplantation. Recent studies have used new techniques to focus on the interplay of thymopoiesis and peripheral expansion that defines T-cell repopulation. Peripheral expansion, driven by host cytokines and antigenic stimulation, dominates early recovery. However, this expansion is often transient and is characterized by limited repertoire diversity. Renewed thymopoiesis has been found to play a critical role in the recovery of repertoire diversity and stable repopulation. The insights gained into the regulation of these processes may provide new therapies to enhance recovery.
Collapse
Affiliation(s)
- Frances T Hakim
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1907, USA.
| | | |
Collapse
|