1
|
Wang Y, Huang X, Luo G, Xu Y, Deng X, Lin Y, Wang Z, Zhou S, Wang S, Chen H, Tao T, He L, Yang L, Yang L, Chen Y, Jin Z, He C, Han Z, Zhang X. The aging lung: microenvironment, mechanisms, and diseases. Front Immunol 2024; 15:1383503. [PMID: 38756780 PMCID: PMC11096524 DOI: 10.3389/fimmu.2024.1383503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/16/2024] [Indexed: 05/18/2024] Open
Abstract
With the development of global social economy and the deepening of the aging population, diseases related to aging have received increasing attention. The pathogenesis of many respiratory diseases remains unclear, and lung aging is an independent risk factor for respiratory diseases. The aging mechanism of the lung may be involved in the occurrence and development of respiratory diseases. Aging-induced immune, oxidative stress, inflammation, and telomere changes can directly induce and promote the occurrence and development of lung aging. Meanwhile, the occurrence of lung aging also further aggravates the immune stress and inflammatory response of respiratory diseases; the two mutually affect each other and promote the development of respiratory diseases. Explaining the mechanism and treatment direction of these respiratory diseases from the perspective of lung aging will be a new idea and research field. This review summarizes the changes in pulmonary microenvironment, metabolic mechanisms, and the progression of respiratory diseases associated with aging.
Collapse
Affiliation(s)
- Yanmei Wang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Xuewen Huang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guofeng Luo
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunying Xu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiqian Deng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhanzhan Wang
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Shuwei Zhou
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Siyu Wang
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Haoran Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Tao
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Lei He
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Luchuan Yang
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Li Yang
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Yutong Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zi Jin
- Department of Anesthesiology and Pain Rehabilitation, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Chengshi He
- Department of Respiratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohong Zhang
- Department of Emergency Medicine Center, Sichuan Province People’s Hospital University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
2
|
Smith AM, Ramirez RM, Harper N, Jimenez F, Branum AP, Meunier JA, Pandranki L, Carrillo A, Winter C, Winter L, Rather CG, Ramirez DA, Andrews CP, Restrepo MI, Maselli DJ, Pugh JA, Clark RA, Lee GC, Moreira AG, Manoharan MS, Okulicz JF, Jacobs RL, Ahuja SK. Large-scale provocation studies identify maladaptive responses to ubiquitous aeroallergens as a correlate of severe allergic rhinoconjunctivitis and asthma. Allergy 2022; 77:1797-1814. [PMID: 34606106 PMCID: PMC9298287 DOI: 10.1111/all.15124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/24/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Allergic asthma (AA) and allergic rhinoconjunctivitis (ARC) are common comorbid environmentally triggered diseases. We hypothesized that severe AA/ARC reflects a maladaptive or unrestrained response to ubiquitous aeroallergens. METHODS We performed provocation studies wherein six separate cohorts of persons (total n = 217) with ARC, with or without AA, were challenged once or more with fixed concentrations of seasonal or perennial aeroallergens in an aeroallergen challenge chamber (ACC). RESULTS Aeroallergen challenges elicited fully or partially restrained vs. unrestrained evoked symptom responsiveness, corresponding to the resilient and adaptive vs. maladaptive AA/ARC phenotypes, respectively. The maladaptive phenotype was evoked more commonly during challenge with a non-endemic versus endemic seasonal aeroallergen. In an AA cohort, symptom responses evoked after house dust mite (HDM) challenges vs. recorded in the natural environment were more accurate and precise predictors of asthma severity and control, lung function (FEV1), and mechanistic correlates of maladaptation. Correlates included elevated levels of peripheral blood CD4+ and CD8+ T-cells, eosinophils, and T-cell activation, as well as gene expression proxies for ineffectual epithelial injury/repair responses. Evoked symptom severity after HDM challenge appeared to be more closely related to levels of CD4+ and CD8+ T-cells than eosinophils, neutrophils, or HDM-specific IgE. CONCLUSIONS Provocation studies support the concept that resilience, adaptation, and maladaptation to environmental disease triggers calibrate AA/ARC severity. Despite the ubiquity of aeroallergens, in response to these disease triggers in controlled settings (ie, ACC), most atopic persons manifest the resilient or adaptive phenotype. Thus, ARC/AA disease progression may reflect the failure to preserve the resilient or adaptive phenotype. The triangulation of CD8+ T-cell activation, airway epithelial injury/repair processes and maladaptation in mediating AA disease severity needs more investigation.
Collapse
|
3
|
Bolivar-Wagers S, Larson JH, Jin S, Blazar BR. Cytolytic CD4 + and CD8 + Regulatory T-Cells and Implications for Developing Immunotherapies to Combat Graft-Versus-Host Disease. Front Immunol 2022; 13:864748. [PMID: 35493508 PMCID: PMC9040077 DOI: 10.3389/fimmu.2022.864748] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/16/2022] [Indexed: 02/03/2023] Open
Abstract
Regulatory T-cells (Treg) are critical for the maintenance of immune homeostasis and tolerance induction. While the immunosuppressive mechanisms of Treg have been extensively investigated for decades, the mechanisms responsible for Treg cytotoxicity and their therapeutic potential in regulating immune responses have been incompletely explored and exploited. Conventional cytotoxic T effector cells (Teffs) are known to be important for adaptive immune responses, particularly in the settings of viral infections and cancer. CD4+ and CD8+ Treg subsets may also share similar cytotoxic properties with conventional Teffs. Cytotoxic effector Treg (cyTreg) are a heterogeneous population in the periphery that retain the capacity to suppress T-cell proliferation and activation, induce cellular apoptosis, and migrate to tissues to ensure immune homeostasis. The latter can occur through several cytolytic mechanisms, including the Granzyme/Perforin and Fas/FasL signaling pathways. This review focuses on the current knowledge and recent advances in our understanding of cyTreg and their potential application in the treatment of human disease, particularly Graft-versus-Host Disease (GVHD).
Collapse
Affiliation(s)
| | | | | | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
4
|
Jiang Y, Deng S, Hu X, Luo L, Zhang Y, Zhang D, Li X, Feng J. Identification of potential biomarkers and immune infiltration characteristics in severe asthma. Int J Immunopathol Pharmacol 2022; 36:3946320221114194. [PMID: 35817495 PMCID: PMC9280849 DOI: 10.1177/03946320221114194] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES We hope to identify key molecules that can be used as markers of asthma severity and investigate their correlation with immune cell infiltration in severe asthma. METHODS An asthma dataset was downloaded from the Gene Expression Omnibus database and then processed by R software to obtain differentially expressed genes (DEGs). First, multiple enrichment platforms were applied to analyze crucial biological processes and pathways and protein-protein interaction networks related to the DEGs. We next combined least absolute shrinkage and selection operator logistic regression and the support vector machine-recursive feature elimination algorithms to screen diagnostic markers of severe asthma. Then, a local cohort consisting of 40 asthmatic subjects (24 with moderate asthma and 16 with severe asthma) was used for biomarker validation. Finally, infiltration of immune cells in asthma bronchoalveolar lavage fluid and their correlation with the screened markers was evaluated by CIBERSORT. RESULTS A total of 97 DEGs were identified in this study. Most of these genes are enriched in T cell activation and immune response in the asthma biological process. CC-chemokine receptor 7 (CCR7) and natural killer cell protein 7(NKG7) were identified as markers of severe asthma. The highest area under the ROC curve (AUC) was from a new indicator combining CCR7 and NKG7 (AUC = 0.851, adj. p < 0.05). Resting and activated memory CD4 T cells, activated NK cells, and CD8 T cells were found to be significantly higher in the severe asthma group (adj. p < 0.01). CCR7 and NKG7 were significantly correlated with these infiltrated cells that showed differences between the two groups. In addition, CCR7 was found to be significantly positively correlated with eosinophils (r = 0.38, adj. p < 0.05) infiltrated in bronchoalveolar lavage fluid. CONCLUSION CCR7 and NKG7 might be used as potential markers for asthma severity, and their expression may be associated with differences in immune cell infiltration in the moderate and severe asthma groups.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Shuanglinzi Deng
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Xinyue Hu
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Lisha Luo
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Yingyu Zhang
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Daimo Zhang
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Xiaozhao Li
- Department of Nephrology, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Juntao Feng
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| |
Collapse
|
5
|
Dey S, Eapen MS, Chia C, Gaikwad AV, Wark PAB, Sohal SS. Pathogenesis, clinical features of asthma COPD overlap (ACO), and therapeutic modalities. Am J Physiol Lung Cell Mol Physiol 2021; 322:L64-L83. [PMID: 34668439 DOI: 10.1152/ajplung.00121.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Both asthma and COPD are heterogeneous diseases identified by characteristic symptoms and functional abnormalities, with airway obstruction common in both diseases. Asthma COPD overlap (ACO) does not define a single disease but is a descriptive term for clinical use that includes several overlapping clinical phenotypes of chronic airways disease with different underlying mechanisms. This literature review was initiated to describe published studies, identify gaps in knowledge, and propose future research goals regarding the disease pathology of ACO, especially the airway remodelling changes and inflammation aspects. Airway remodelling occurs in asthma and COPD, but there are differences in the structures affected and the prime anatomic site at which they occur. Reticular basement membrane thickening and cellular infiltration with eosinophils and T-helper (CD4+) lymphocytes are prominent features of asthma. Epithelial squamous metaplasia, airway wall fibrosis, emphysema, bronchoalveolar lavage (BAL) neutrophilia and (CD8+) T-cytotoxic lymphocyte infiltrations in the airway wall are features of COPD. There is no universally accepted definition of ACO, nor are there clearly defined pathological characteristics to differentiate from asthma and COPD. Understanding etiological concepts within the purview of inflammation and airway remodelling changes in ACO would allow better management of these patients.
Collapse
Affiliation(s)
- Surajit Dey
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Mathew Suji Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Collin Chia
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia.,Department of Respiratory Medicine, Launceston General Hospital, Launceston, Tasmania, Australia
| | - Archana Vijay Gaikwad
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia.,Department of Respiratory and Sleep Medicine John Hunter Hospital, New Lambton Heights, Australia
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| |
Collapse
|
6
|
Schuliga M, Read J, Knight DA. Ageing mechanisms that contribute to tissue remodeling in lung disease. Ageing Res Rev 2021; 70:101405. [PMID: 34242806 DOI: 10.1016/j.arr.2021.101405] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/13/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Age is a major risk factor for chronic respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and certain phenotypes of asthma. The recent COVID-19 pandemic also highlights the increased susceptibility of the elderly to acute respiratory distress syndrome (ARDS), a diffuse inflammatory lung injury with often long-term effects (ie parenchymal fibrosis). Collectively, these lung conditions are characterized by a pathogenic reparative process that, rather than restoring organ function, contributes to structural and functional tissue decline. In the ageing lung, the homeostatic control of wound healing following challenge or injury has an increased likelihood of being perturbed, increasing susceptibility to disease. This loss of fidelity is a consequence of a diverse range of underlying ageing mechanisms including senescence, mitochondrial dysfunction, proteostatic stress and diminished autophagy that occur within the lung, as well as in other tissues, organs and systems of the body. These ageing pathways are highly interconnected, involving localized and systemic increases in inflammatory mediators and damage associated molecular patterns (DAMPs); along with corresponding changes in immune cell function, metabolism and composition of the pulmonary and gut microbiomes. Here we comprehensively review the roles of ageing mechanisms in the tissue remodeling of lung disease.
Collapse
Affiliation(s)
- Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| | - Jane Read
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Providence Health Care Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Asthma and COVID-19: Emphasis on Adequate Asthma Control. Can Respir J 2021; 2021:9621572. [PMID: 34457096 PMCID: PMC8397565 DOI: 10.1155/2021/9621572] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/18/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Asthmatics are at an increased risk of developing exacerbations after being infected by respiratory viruses such as influenza virus, parainfluenza virus, and human and severe acute respiratory syndrome coronaviruses (SARS-CoV). Asthma, especially when poorly controlled, is an independent risk factor for developing pneumonia. A subset of asthmatics can have significant defects in their innate, humoral, and cell-mediated immunity arms, which may explain the increased susceptibility to infections. Adequate asthma control is associated with a significant decrease in episodes of exacerbation. Because of their wide availability and potency to promote adequate asthma control, glucocorticoids, especially inhaled ones, are the cornerstone of asthma management. The current COVID-19 pandemic affects millions of people worldwide and possesses mortality several times that of seasonal influenza; therefore, it is necessary to revisit this subject. The pathogenesis of SARS-CoV-2, the virus that causes COVID-19, can potentiate the development of acute asthmatic exacerbation with the potential to worsen the state of chronic airway inflammation. The relationship is evident from several studies that show asthmatics experiencing a more adverse clinical course of SARS-CoV-2 infection than nonasthmatics. Recent studies show that dexamethasone, a potent glucocorticoid, and other inhaled corticosteroids significantly reduce morbidity and mortality among hospitalized COVID-19 patients. Hence, while we are waiting for more studies with higher level of evidence that further narrate the association between COVID-19 and asthma, we advise clinicians to try to achieve adequate disease control in asthmatics as it may reduce incidences and severity of exacerbations especially from SARS-CoV-2 infection.
Collapse
|
8
|
Gelfand EW, Joetham A, Wang M, Takeda K, Schedel M. Spectrum of T-lymphocyte activities regulating allergic lung inflammation. Immunol Rev 2017; 278:63-86. [PMID: 28658551 PMCID: PMC5501488 DOI: 10.1111/imr.12561] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite advances in the treatment of asthma, optimization of symptom control remains an unmet need in many patients. These patients, labeled severe asthma, are responsible for a substantial fraction of the disease burden. In these patients, research is needed to define the cellular and molecular pathways contributing to disease which in large part are refractory to corticosteroid treatment. The causes of steroid-resistant asthma are multifactorial and result from complex interactions of genetics, environmental factors, and innate and adaptive immunity. Adaptive immunity, addressed here, integrates the activities of distinct T-cell subsets and by definition is dynamic and responsive to an ever-changing environment and the influences of epigenetic modifications. These T-cell subsets exhibit different susceptibilities to the actions of corticosteroids and, in some, corticosteroids enhance their functional activation. Moreover, these subsets are not fixed in lineage differentiation but can undergo transcriptional reprogramming in a bidirectional manner between protective and pathogenic effector states. Together, these factors contribute to asthma heterogeneity between patients but also in the same patient at different stages of their disease. Only by carefully defining mechanistic pathways, delineating their sensitivity to corticosteroids, and determining the balance between regulatory and effector pathways will precision medicine become a reality with selective and effective application of targeted therapies.
Collapse
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Anthony Joetham
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Meiqin Wang
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Katsuyuki Takeda
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| |
Collapse
|
9
|
Li P, Yang QZ, Wang W, Zhang GQ, Yang J. Increased IL-4- and IL-17-producing CD8 + cells are related to decreased CD39 +CD4 +Foxp3 + cells in allergic asthma. J Asthma 2017; 55:8-14. [PMID: 28346024 DOI: 10.1080/02770903.2017.1310225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE In allergic asthma, regulatory T cell (Treg) number and function are decreased. Antigen-primed CD8+ T cells play an indispensable role in the full development of airway inflammation and airway hyper-responsiveness (AHR) occurring in asthma. In this study, we investigated the relationship between subpopulations of CD8+ T cells and CD39+ Tregs. METHODS Female C57BL/6 mice were used to develop the model of allergic asthma. Experimental mice were immunized with ovalbumin (OVA) by intra-peritoneal (i.p) injection and then challenged with OVA by intra-tracheal administration. Control mice were immunized with vehicle by i.p injection and challenged with OVA. Airway inflammation was determined by histology and AHR was measured by an invasive method. Levels of interferon (IFN)-γ, IL-4, and IL-17 in bronchoalveolar lavage fluid (BALF) were determined by enzyme-linked immunosorbent assay. The frequencies of CD8+IFN-γ+ cells (Tc1), CD8+IL-4+ cells (Tc2), CD8+IL-17+cells (Tc17), and CD39+Tregs were measured by flow cytometry. The correlation between CD39+Tregs and Tc subsets was analyzed by Pearson's test. RESULTS Experimental mice displayed phenotypes of allergic asthma, including inflammatory cell infiltration into the lungs, goblet cell hyperplasia, increased airway resistance, and increased IL-4 and IL-17 in BALF. Compared to control mice, experimental mice displayed lower CD39+Tregs and Tc1 but higher Tc2 and Tc17. There was a negative correlation between CD39+Tregs and Tc2 or Tc17. CONCLUSION In allergic asthma, increased Tc2 and Tc17 are possibly related to insufficient CD39+Tregs.
Collapse
Affiliation(s)
- Ping Li
- a Department of Pulmonology , Affiliated Xiangyang Hospital of Hubei University of Medicine , Xiangyang , China
| | - Qun-Zhen Yang
- a Department of Pulmonology , Affiliated Xiangyang Hospital of Hubei University of Medicine , Xiangyang , China
| | - Wei Wang
- b Department of Pulmonology , Affiliated Zhongnan Hospital of Wuhan University , Wuhan , China
| | - Gu-Qin Zhang
- b Department of Pulmonology , Affiliated Zhongnan Hospital of Wuhan University , Wuhan , China
| | - Jiong Yang
- b Department of Pulmonology , Affiliated Zhongnan Hospital of Wuhan University , Wuhan , China
| |
Collapse
|
10
|
Lourenço O, Fonseca AM, Taborda-Barata L. Human CD8+ T Cells in Asthma: Possible Pathways and Roles for NK-Like Subtypes. Front Immunol 2016; 7:638. [PMID: 28066445 PMCID: PMC5179570 DOI: 10.3389/fimmu.2016.00638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/12/2016] [Indexed: 12/22/2022] Open
Abstract
Asthma affects approximately 300 million people worldwide and is the most common chronic lung disease, which usually is associated with bronchial inflammation. Most research has focused upon the role of CD4+ T cells, and relatively few studies have addressed the phenotypic and functional roles of CD8+ T cell types and subtypes. Human NK-like CD8+ T cells may involve cells that have been described as CD8+CD28−, CD8+CD28−CD57+, CD8+CD27−, or CD8+ effector memory (TEM) cells, among other. However, most of the data that are available regarding these various cell types were obtained in murine models did not thoroughly characterize these cells with phenotypically or functionally or did not involve asthma-related settings. Nevertheless, one may conceptualize three principal roles for human NK-like CD8+ T cells in asthma: disease-promoting, regulatory, and/or tissue repair. Although evidence for some of these roles is scarce, it is possible to extrapolate some data from overlapping or related CD8+ T cell phenotypes, with caution. Clearly, further research is warranted, namely in terms of thorough functional and phenotypic characterization of human NK-like CD8+ T cells in human asthma of varying severity.
Collapse
Affiliation(s)
- Olga Lourenço
- CICS - UBI, Health Sciences Research Centre, University of Beira Interior , Covilhã , Portugal
| | - Ana Mafalda Fonseca
- CICS - UBI, Health Sciences Research Centre, University of Beira Interior , Covilhã , Portugal
| | - Luis Taborda-Barata
- CICS - UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal; Department of Allergy and Clinical Immunology, Cova da Beira Hospital Centre, Covilhã, Portugal
| |
Collapse
|
11
|
Michel JJ, Griffin P, Vallejo AN. Functionally Diverse NK-Like T Cells Are Effectors and Predictors of Successful Aging. Front Immunol 2016; 7:530. [PMID: 27933066 PMCID: PMC5121286 DOI: 10.3389/fimmu.2016.00530] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022] Open
Abstract
The fundamental challenge of aging and long-term survivorship is maintenance of functional independence and compression of morbidity despite a life history of disease. Inasmuch as immunity is a determinant of individual health and fitness, unraveling novel mechanisms of immune homeostasis in late life is of paramount interest. Comparative studies of young and old persons have documented age-related atrophy of the thymus, the contraction of diversity of the T cell receptor (TCR) repertoire, and the intrinsic inefficiency of classical TCR signaling in aged T cells. However, the elderly have highly heterogeneous health phenotypes. Studies of defined populations of persons aged 75 and older have led to the recognition of successful aging, a distinct physiologic construct characterized by high physical and cognitive functioning without measurable disability. Significantly, successful agers have a unique T cell repertoire; namely, the dominance of highly oligoclonal αβT cells expressing a diverse array of receptors normally expressed by NK cells. Despite their properties of cell senescence, these unusual NK-like T cells are functionally active effectors that do not require engagement of their clonotypic TCR. Thus, NK-like T cells represent a beneficial remodeling of the immune repertoire with advancing age, consistent with the concept of immune plasticity. Significantly, certain subsets are predictors of physical/cognitive performance among older adults. Further understanding of the roles of these NK-like T cells to host defense, and how they integrate with other physiologic domains of function are new frontiers for investigation in Aging Biology. Such pursuits will require a research paradigm shift from the usual young-versus-old comparison to the analysis of defined elderly populations. These endeavors may also pave way to age-appropriate, group-targeted immune interventions for the growing elderly population.
Collapse
Affiliation(s)
- Joshua J Michel
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Patricia Griffin
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Abbe N Vallejo
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Pittsburgh Claude Pepper Older Americans Independence Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Hodge S, Hodge G, Simpson JL, Yang IA, Upham J, James A, Gibson PG, Reynolds PN. Blood cytotoxic/inflammatory mediators in non-eosinophilic asthma. Clin Exp Allergy 2016; 46:60-70. [PMID: 26767492 DOI: 10.1111/cea.12634] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 08/31/2015] [Accepted: 09/03/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND Non-eosinophilic asthma (NEA) is a distinct, often corticosteroid-resistant inflammatory asthma phenotype. NK and NKT-like cells are effector lymphocytes that we have shown, like CD28null T cells, to be relatively resistant to steroids and major sources of pro-inflammatory/cytotoxic mediators. We hypothesized that these cells and mediators would be increased in peripheral blood in NEA. METHODS Adults with severe asthma and variable airflow obstruction, poorly controlled despite maintenance therapy with inhaled glucocorticosteroids and long-acting bronchodilators, were recruited. Blood was assessed in those with eosinophilic asthma (n = 12), NEA (n = 25) and healthy non-smoking controls (n = 30). We applied flow cytometry to measure T, CD28null, NK and NKT-like cells and their expression of granzyme B, perforin, and killer inhibitory/activating receptors CD94(Kp43), CD158b and CD107A. Intracellular pro-inflammatory cytokine production (IFN-γ and TNF-α) was assessed in 18 controls and 10 patients with asthma/group. RESULTS In NEA, there was increased expression of granzyme B by CD8+ T cells vs. CONTROLS There was increased expression of granzyme B and CD158 and decreased CD94 on NK cells, vs. healthy controls and those with eosinophilic asthma. IFN-γ production by NK cells and TNF-α production by NKT-like cells in NEA were significantly increased vs. CONTROLS In both eosinophilic and NEA phenotypes, there were significant increases in CD4+28null T cells (72% and 81% increases, respectively, vs. controls) and their expression of pro-inflammatory cytokines. Significant correlations were noted between blood CD4+28null T cells and neutrophil numbers in induced sputum, and between corticosteroid dose and blood NKT-like cells, and their production of granzyme B and TNF-α and NK IFN-γ. CONCLUSION AND CLINICAL RELEVANCE In poorly controlled asthma, altered expression of cytotoxic/pro-inflammatory mediators can be seen on a variety of lymphocyte subsets in the peripheral blood; these changes are most apparent in NEA. Whether this pattern of expression is a marker of treatment responsiveness and future risk of exacerbations remains to be determined.
Collapse
Affiliation(s)
- S Hodge
- Lung Research, Hanson Institute and Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia.,Department of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - G Hodge
- Lung Research, Hanson Institute and Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia.,Department of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - J L Simpson
- Centre for Asthma and Respiratory Disease, The University of Newcastle, Newcastle, NSW, Australia.,Department of Respiratory and Sleep Medicine, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - I A Yang
- School of Medicine, The University of Queensland, Brisbane, QLD, Australia.,The Prince Charles Hospital, Brisbane, QLD, Australia
| | - J Upham
- School of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - A James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - P G Gibson
- Centre for Asthma and Respiratory Disease, The University of Newcastle, Newcastle, NSW, Australia.,Department of Respiratory and Sleep Medicine, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - P N Reynolds
- Lung Research, Hanson Institute and Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia.,Department of Medicine, University of Adelaide, Adelaide, SA, Australia
| | | |
Collapse
|
13
|
Huber M, Lohoff M. Change of paradigm: CD8+ T cells as important helper for CD4+ T cells during asthma and autoimmune encephalomyelitis. ACTA ACUST UNITED AC 2015; 24:8-15. [PMID: 26120542 PMCID: PMC4479451 DOI: 10.1007/s40629-015-0038-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/13/2014] [Indexed: 12/24/2022]
Abstract
The activation of naive CD4+ and CD8+ T cells in response to antigen and their subsequent proliferation and differentiation into effectors are important features of a cell-mediated immune response. CD4+ T cells (also known as T helper cells, Th) differentiate into several subpopulations including Th1, Th2, Th9, Th17, Tfh and Treg cells, characterized by specific cytokine profiles and effector functions. However, recent evidence indicates that CD8+ T cells (termed cytotoxic T lymphocytes, CTLs or Tc cells) can differentiate into subpopulations with similar characteristics denoted as Tc2, Tc9, Tc17 and CD8+ Treg cells in addition to CTLs. Although these subpopulations accomplish important protective functions, their uncontrolled responses cause immunopathology including allergy and autoimmunity. Our recent findings indicate a change of paradigm: during these pathologic responses, CD8+ T cell subpopulations act as strong helpers for the activity of CD4+ T cells rather than being cytotoxic. In this review, we focus on the role of Th2, Th9, Th17 as well as Tc9 and Tc17 cells in asthma and autoimmune encephalomyelitis and on their interaction during these immunopathologic responses. Cite this as Huber M, Lohoff M. Change of paradigm: CD8+ T cells as important helper for CD4+ T cells during asthma and autoimmune encephalomyelitis. Allergo J Int 2015;24:8–15 DOI: 10.1007/s40629-015-0038-4
Collapse
Affiliation(s)
- Magdalena Huber
- />Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
- />Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Hans-Meerwein-Straße 2, 35032 Marburg, Germany
| | - Michael Lohoff
- />Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| |
Collapse
|
14
|
Ein Paradigmenwechsel: CD8+ T-Zellen als wichtige Helfer für CD4+ T-Zellen während Asthma und autoimmuner Encephalomyelitis. ALLERGO JOURNAL 2015. [DOI: 10.1007/s15007-015-0751-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Engela AU, Baan CC, Litjens NHR, Franquesa M, Betjes MGH, Weimar W, Hoogduijn MJ. Mesenchymal stem cells control alloreactive CD8(+) CD28(-) T cells. Clin Exp Immunol 2014; 174:449-58. [PMID: 24028656 DOI: 10.1111/cei.12199] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2013] [Indexed: 12/18/2022] Open
Abstract
CD28/B7 co-stimulation blockade with belatacept prevents alloreactivity in kidney transplant patients. However, cells lacking CD28 are not susceptible to belatacept treatment. As CD8(+) CD28(-) T-cells have cytotoxic and pathogenic properties, we investigated whether mesenchymal stem cells (MSC) are effective in controlling these cells. In mixed lymphocyte reactions (MLR), MSC and belatacept inhibited peripheral blood mononuclear cell (PBMC) proliferation in a dose-dependent manner. MSC at MSC/effector cell ratios of 1:160 and 1:2·5 reduced proliferation by 38·8 and 92·2%, respectively. Belatacept concentrations of 0·1 μg/ml and 10 μg/ml suppressed proliferation by 20·7 and 80·6%, respectively. Both treatments in combination did not inhibit each other's function. Allostimulated CD8(+) CD28(-) T cells were able to proliferate and expressed the cytolytic and cytotoxic effector molecules granzyme B, interferon (IFN)-γ and tumour necrosis factor (TNF)-α. While belatacept did not affect the proliferation of CD8(+) CD28(-) T cells, MSC reduced the percentage of CD28(-) T cells in the proliferating CD8(+) T cell fraction by 45·9% (P = 0·009). CD8(+) CD28(-) T cells as effector cells in MLR in the presence of CD4(+) T cell help gained CD28 expression, an effect independent of MSC. In contrast, allostimulated CD28(+) T cells did not lose CD28 expression in MLR-MSC co-culture, suggesting that MSC control pre-existing CD28(-) T cells and not newly induced CD28(-) T cells. In conclusion, alloreactive CD8(+) CD28(-) T cells that remain unaffected by belatacept treatment are inhibited by MSC. This study indicates the potential of an MSC-belatacept combination therapy to control alloreactivity.
Collapse
Affiliation(s)
- A U Engela
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
16
|
Proklou A, Soulitzis N, Neofytou E, Rovina N, Zervas E, Gaga M, Siafakas NM, Tzortzaki EG. Granule Cytotoxic Activity and Oxidative DNA Damage in Smoking and Nonsmoking Patients With Asthma. Chest 2013; 144:1230-1237. [DOI: 10.1378/chest.13-0367] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
17
|
Hodge G, Hodge S, Ahern J, Holmes-Liew CL, Reynolds PN, Holmes M. Up-regulation of alternate co-stimulatory molecules on proinflammatory CD28null T cells in bronchiolitis obliterans syndrome. Clin Exp Immunol 2013; 173:150-60. [PMID: 23607447 DOI: 10.1111/cei.12081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2013] [Indexed: 12/01/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is associated with lack of immunosuppression of T cell proinflammatory cytokines and increased T cell granzyme B. Repeated antigen-driven proliferation down-regulates T cell CD28. We hypothesized that down-regulation of CD28 and up-regulation of alternate co-stimulatory molecules (CD134, CD137, CD152 and CD154) on T cells may be associated with BOS. Co-stimulatory molecules, granzyme B, perforin and intracellular cytokines were measured by flow cytometry on T cells from stable lung transplant patients (n = 38), patients with BOS (n = 20) and healthy controls (n = 10). There was a significant increase in the percentage of CD4/28(null) and CD8/28(null) T cells producing granzyme B, interferon (IFN)-γ and tumour necrosis factor (TNF)-α in BOS compared with stable patients. Down-regulation of CD28 was associated with steroid resistance and up-regulation of CD134, CD137, CD152 and CD154 on CD4(+) T cells and CD137 and CD152 on CD8(+) T cells. There was a significant correlation between increased CD28(null) /CD137 T cells producing IFN-γ, TNF-α with BOS grade (r = 0·861, P < 0·001 for CD28(null) /CD137 IFN-γ/CD8) and time post-transplant (r = 0·698, P < 0·001 for CD28(null) /CD137 IFN-γ/CD8). BOS is associated with down-regulation of CD28 and up-regulation of alternate co-stimulatory molecules on steroid-resistant peripheral blood proinflammatory CD4(+) and CD8(+) T cells. Therapeutic targeting of alternate co-stimulatory molecules on peripheral blood CD28(null) T cells and monitoring response using these assays may help in the management of patients with BOS.
Collapse
Affiliation(s)
- G Hodge
- Lung Research, Hanson Institute and Department of Thoracic Medicine, Royal Adelaide Hospital, Australia.
| | | | | | | | | | | |
Collapse
|
18
|
Signification clinique des expansions polyclonales lymphocytaires T CD8+/CD57+. Presse Med 2013; 42:327-37. [DOI: 10.1016/j.lpm.2012.04.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 04/19/2012] [Accepted: 04/25/2012] [Indexed: 12/27/2022] Open
|
19
|
Visekruna A, Ritter J, Scholz T, Campos L, Guralnik A, Poncette L, Raifer H, Hagner S, Garn H, Staudt V, Bopp T, Reuter S, Taube C, Loser K, Huber M. Tc9 cells, a new subset of CD8(+) T cells, support Th2-mediated airway inflammation. Eur J Immunol 2013; 43:606-18. [PMID: 23254356 DOI: 10.1002/eji.201242825] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 12/07/2012] [Accepted: 12/13/2012] [Indexed: 12/29/2022]
Abstract
Similar to T-helper (Th) cells, CD8(+) T cells also differentiate into distinct subpopulations. However, the existence of IL-9-producing CD8(+) T (Tc9) cells has not been elucidated so far. We show that murine CD8(+) T cells activated in the presence of IL-4 plus TGF-β develop into transient IL-9 producers characterized by specific IFN-γ and IL-10 expression patterns as well as by low cytotoxic function along with diminished expression of the CTL-associated transcription factors T-bet and Eomesodermin. Similarly to the CD4(+) counterpart, Tc9 cells required for their differentiation STAT6 and IRF4. Tc9 cells deficient for these master regulators displayed increased levels of Foxp3 that in turn suppressed IL-9 production. In an allergic airway disease model, Tc9 cells promoted the onset of airway inflammation, mediated by subpathogenic numbers of Th2 cells. This support was specific for Tc9 cells because CTLs failed to exert this function. We detected increased Tc9 frequency in the periphery in mice and humans with atopic dermatitis, a Th2-associated skin disease that often precedes asthma. Thus, our data point to the existence of Tc9 cells and to their supportive function in Th2-dependent airway inflammation, suggesting that these cells might be a therapeutic target in allergic disorders.
Collapse
Affiliation(s)
- Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, University of Marburg, Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Tsitsiou E, Williams AE, Moschos SA, Patel K, Rossios C, Jiang X, Adams OD, Macedo P, Booton R, Gibeon D, Chung KF, Lindsay MA. Transcriptome analysis shows activation of circulating CD8+ T cells in patients with severe asthma. J Allergy Clin Immunol 2011; 129:95-103. [PMID: 21917308 DOI: 10.1016/j.jaci.2011.08.011] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 08/08/2011] [Accepted: 08/12/2011] [Indexed: 10/17/2022]
Abstract
BACKGROUND Although previous studies have implicated tissue CD4(+) T cells in the development and maintenance of the inflammatory response in asthmatic patients, little is known about the role of CD8(+) T cells. There is now accumulating evidence that microRNAs and other noncoding RNAs are important regulators of T-cell function. OBJECTIVES We sought to use transcriptomics to determine the activation state of circulating CD4(+) and CD8(+) T cells in patients with nonsevere and severe asthma. METHODS mRNA and noncoding RNA expression in circulating T cells was measured by means of microarray, quantitative real-time PCR, or both. RESULTS Comparison of mRNA expression showed widespread changes in the circulating CD8(+) but not CD4(+) T cells from patients with severe asthma. No changes were observed in the CD4(+) and CD8(+) T cells in patients with nonsevere asthma versus those in healthy control subjects. Bioinformatics analysis showed that the changes in CD8(+) T-cell mRNA expression were associated with multiple pathways involved in T-cell activation. As with mRNAs, we also observed widespread changes in expression of noncoding RNA species, including natural antisense, pseudogenes, intronic long noncoding RNAs (lncRNAs), and intergenic lncRNAs in CD8(+) T cells from patients with severe asthma. Measurement of the microRNA expression profile showed selective downregulation of miR-28-5p in CD8(+) T cells and reduction of miR-146a and miR-146b in both CD4(+) and CD8(+) T cells. CONCLUSIONS Severe asthma is associated with the activation of circulating CD8(+) T cells but not CD4(+) T cells. This response is correlated with the downregulation of miR-146a/b and miR-28-5p, as well as changes in the expression of multiple species of lncRNA that might regulate CD8(+) T-cell function.
Collapse
Affiliation(s)
- Eleni Tsitsiou
- Respiratory Research Group, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Strioga M, Pasukoniene V, Characiejus D. CD8+ CD28- and CD8+ CD57+ T cells and their role in health and disease. Immunology 2011; 134:17-32. [PMID: 21711350 DOI: 10.1111/j.1365-2567.2011.03470.x] [Citation(s) in RCA: 354] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic antigenic stimulation leads to gradual accumulation of late-differentiated, antigen-specific, oligoclonal T cells, particularly within the CD8(+) T-cell compartment. They are characterized by critically shortened telomeres, loss of CD28 and/or gain of CD57 expression and are defined as either CD8(+) CD28(-) or CD8(+) CD57(+) T lymphocytes. There is growing evidence that the CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell population plays a significant role in various diseases or conditions, associated with chronic immune activation such as cancer, chronic intracellular infections, chronic alcoholism, some chronic pulmonary diseases, autoimmune diseases, allogeneic transplantation, as well as has a great influence on age-related changes in the immune system status. CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell population is heterogeneous and composed of various functionally competing (cytotoxic and immunosuppressive) subsets thus the overall effect of CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell-mediated immunity depends on the predominance of a particular subset. Many articles claim that CD8(+) CD28(-) (CD8(+) CD57(+)) T cells have lost their proliferative capacity during process of replicative senescence triggered by repeated antigenic stimulation. However recent data indicate that CD8(+) CD28(-) (CD8(+) CD57(+)) T cells can transiently up-regulate telomerase activity and proliferate under certain stimulation conditions. Similarly, conflicting data is provided regarding CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell sensitivity to apoptosis, finally leading to the conclusion that this T-cell population is also heterogeneous in terms of its apoptotic potential. This review provides a comprehensive approach to the CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell population: we describe in detail its origins, molecular and functional characteristics, subsets, role in various diseases or conditions, associated with persistent antigenic stimulation.
Collapse
Affiliation(s)
- Marius Strioga
- Laboratory of Immunology, Institute of Oncology, Vilnius University, Vilnius Faculty of Medicine, Vilnius University, Vilnius, Lithuania.
| | | | | |
Collapse
|
22
|
Madore AM, Laprise C. Immunological and genetic aspects of asthma and allergy. J Asthma Allergy 2010; 3:107-21. [PMID: 21437045 PMCID: PMC3047903 DOI: 10.2147/jaa.s8970] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Indexed: 12/21/2022] Open
Abstract
Prevalence of allergy and allergic asthma are increasing worldwide. More than half of the US population has a positive skin prick test and approximately 10% are asthmatics. Many studies have been conducted to define immunological pathways underlying allergy and asthma development and to identify the main genetic determinants. In the effort to find missing pieces of the puzzle, new genomic approaches and more standardized ones, such as the candidate gene approach, have been used collectively. This article proposes an overview of the actual knowledge about immunological and genetic aspects of allergy and asthma. Special attention has been drawn to the challenges linked to genetic research in complex traits such as asthma and to the contribution of new genomic approaches.
Collapse
Affiliation(s)
- Anne-Marie Madore
- Université du Québec à Chicoutimi, Département des sciences fondamentales, Saguenay, Canada
| | | |
Collapse
|
23
|
Lourenço O, Mafalda Fonseca A, Taborda-Barata L. T cells in sputum of asthmatic patients are activated independently of disease severity or control. Allergol Immunopathol (Madr) 2009; 37:285-92. [PMID: 19850398 DOI: 10.1016/j.aller.2009.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 05/06/2009] [Accepted: 05/08/2009] [Indexed: 11/24/2022]
Abstract
BACKGROUND T cells play an important role in bronchial asthma. Although airway CD4+ T cells have been extensively studied previously, there are hardly any studies relating CD8+ T cell activation and disease symptoms. OBJECTIVES The aim of this study was to analyse the association between T cell activation in induced sputum T cells and asthma severity and control; and to evaluate T cell subpopulations in the same subgroups. METHODS Fifty allergic asthmatic patients were recruited and lung function testing was performed. Airway cells were obtained by sputum induction via inhalation of hypertonic saline solution. CD3, CD4, CD8, CD28, CD25 and CD69 were studied by flow cytometry in whole induced sputum and peripheral blood cells. RESULTS Total induced sputum T cells and CD8+ T cells had a higher relative percentage of the activation markers CD25 and CD69 in comparison with peripheral blood. In sputum, the relative percentage of CD25 was higher in CD4+ T cells when compared to CD8+ T cells and the reverse was true regarding CD69. However, neither disease severity nor control were associated with the relative percentage of CD25 or CD69 expression on T cells in sputum. CONCLUSIONS Both CD4+ and CD8+ T cells are activated in the lungs and peripheral blood of asthmatic patients. However, with the possible exception of CD69+ CD8+ T lymphocytes in the sputum, there is no association between T cell activation phenotype in the target organ and disease severity or control.
Collapse
|
24
|
|
25
|
Bernstein IL, Li JT, Bernstein DI, Hamilton R, Spector SL, Tan R, Sicherer S, Golden DBK, Khan DA, Nicklas RA, Portnoy JM, Blessing-Moore J, Cox L, Lang DM, Oppenheimer J, Randolph CC, Schuller DE, Tilles SA, Wallace DV, Levetin E, Weber R. Allergy diagnostic testing: an updated practice parameter. Ann Allergy Asthma Immunol 2008; 100:S1-148. [PMID: 18431959 DOI: 10.1016/s1081-1206(10)60305-5] [Citation(s) in RCA: 291] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
26
|
Hamzaoui A, Cheik Rouhou S, Graïri H, Abid H, Ammar J, Chelbi H, Hamzaoui K. NKT cells in the induced sputum of severe asthmatics. Mediators Inflamm 2007; 2006:71214. [PMID: 16883065 PMCID: PMC1592585 DOI: 10.1155/mi/2006/71214] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
To determine whether there was a specific inflammatory process in
severe asthmatics, the phenotypic characteristics of induced
sputum immune cells were analysed among patients with severe
asthma. Twenty-two induced sputa (10 severe asthmatics) were
studied. Flow cytometric analysis was performed using immune cells
of the sputum and monoclonal antibodies to CD3, CD4, CD8, CD56,
CD25, and TCRγδ. The number of NKT
(CD3+CD56+) cells was significantly higher in the sputum
of severe asthmatics compared with mild asthmatic and
healthy control groups (P < .05). CD8+CD56+ cells were
the predominant subtype of the increased NKT cells in severe
asthmatics. CD3+CD56+Vα24+,
TCRγδ+ CD56+, and CD4+CD25+ T cells were
significantly increased in severe asthmatic patients. These
results suggest that the immunopathogenesis of severe asthmatics
vary between severe and mild asthmatics, and that
CD8+CD56+ NKT cells may play an important role in the
immunopathogenesis of severe asthma.
Collapse
Affiliation(s)
- Agnes Hamzaoui
- Department of Pediatric and Respiratory Diseases,
Abderrahmane Mami Hospital, Pavillon B, Ariana 2080, Tunisia
- Homeostasis and Cell Dysfunction Unit Research
99/UR/08-40, Medicine University of Tunis, 1007 Tunis, Tunisia
| | - Sana Cheik Rouhou
- Department of Pediatric and Respiratory Diseases,
Abderrahmane Mami Hospital, Pavillon B, Ariana 2080, Tunisia
- Homeostasis and Cell Dysfunction Unit Research
99/UR/08-40, Medicine University of Tunis, 1007 Tunis, Tunisia
| | - Hedia Graïri
- Department of Pediatric and Respiratory Diseases,
Abderrahmane Mami Hospital, Pavillon B, Ariana 2080, Tunisia
- Homeostasis and Cell Dysfunction Unit Research
99/UR/08-40, Medicine University of Tunis, 1007 Tunis, Tunisia
| | - Hanadi Abid
- Department of Pediatric and Respiratory Diseases,
Abderrahmane Mami Hospital, Pavillon B, Ariana 2080, Tunisia
- Homeostasis and Cell Dysfunction Unit Research
99/UR/08-40, Medicine University of Tunis, 1007 Tunis, Tunisia
| | - Jamel Ammar
- Department of Pediatric and Respiratory Diseases,
Abderrahmane Mami Hospital, Pavillon B, Ariana 2080, Tunisia
- Homeostasis and Cell Dysfunction Unit Research
99/UR/08-40, Medicine University of Tunis, 1007 Tunis, Tunisia
| | - Hanene Chelbi
- Homeostasis and Cell Dysfunction Unit Research
99/UR/08-40, Medicine University of Tunis, 1007 Tunis, Tunisia
| | - Kamel Hamzaoui
- Homeostasis and Cell Dysfunction Unit Research
99/UR/08-40, Medicine University of Tunis, 1007 Tunis, Tunisia
- *Kamel Hamzaoui:
| |
Collapse
|