1
|
Miyamoto E, Vosoughi D, Wang J, Al-Refaee J, Berra G, Daigneault T, Duong A, Joe B, Moshkelgosha S, Keshavjee S, Tinckam K, Hwang D, Chruscinski A, Juvet S, Martinu T. Local intragraft humoral immune responses in chronic lung allograft dysfunction. J Heart Lung Transplant 2025; 44:105-117. [PMID: 39097215 DOI: 10.1016/j.healun.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND Donor human leukocyte antigen (HLA)-specific antibodies (DSA) and non-HLA antibodies can cause allograft injury, possibly leading to chronic lung allograft dysfunction (CLAD) after lung transplantation. It remains unclear whether these antibodies are produced locally in the graft or derived solely from circulation. We hypothesized that DSA and non-HLA antibodies are produced in CLAD lungs. METHODS Lung tissue was prospectively collected from 15 CLAD patients undergoing retransplantation or autopsy. 0.3 g of fresh lung tissue was cultured for 4 days without or with lipopolysaccharide or CD40L: lung culture supernatant (LCS) was sampled. Protein eluate was obtained from 0.3 g of frozen lung tissue. The mean fluorescence intensity (MFI) of DSA and non-HLA antibodies was measured by Luminex and antigen microarray, respectively. RESULTS LCS from all 4 patients who had serum DSA at lung isolation were positive for DSA, with higher levels measured after CD40L stimulation (CD40L+LCS). Of these, only 2 had detectable DSA in lung eluate. MFI of non-HLA antibodies from CD40L+LCS correlated with those from lung eluate but not with those from sera. Flow cytometry showed higher frequencies of activated lung B cells in patients whose CD40L+LCS was positive for DSA (n = 4) or high non-HLA antibodies (n = 6) compared to those with low local antibodies (n = 5). Immunofluorescence staining showed CLAD lung lymphoid aggregates with local antibodies contained larger numbers of IgG+ plasma cells and greater IL-21 expression. CONCLUSIONS We show that DSA and non-HLA antibodies can be produced within activated B cell-rich lung allografts.
Collapse
Affiliation(s)
- Ei Miyamoto
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Daniel Vosoughi
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Jinguo Wang
- HLA Laboratory, Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Jamal Al-Refaee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Gregory Berra
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Tina Daigneault
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Allen Duong
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Betty Joe
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Sajad Moshkelgosha
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Kathryn Tinckam
- HLA Laboratory, Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - David Hwang
- Department of Pathology, Sunnybrook Hospital, Toronto, Ontario, Canada
| | | | - Stephen Juvet
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Zhao L, Jin S, Wang S, Zhang Z, Wang X, Chen Z, Wang X, Huang S, Zhang D, Wu H. Tertiary lymphoid structures in diseases: immune mechanisms and therapeutic advances. Signal Transduct Target Ther 2024; 9:225. [PMID: 39198425 PMCID: PMC11358547 DOI: 10.1038/s41392-024-01947-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are defined as lymphoid aggregates formed in non-hematopoietic organs under pathological conditions. Similar to secondary lymphoid organs (SLOs), the formation of TLSs relies on the interaction between lymphoid tissue inducer (LTi) cells and lymphoid tissue organizer (LTo) cells, involving multiple cytokines. Heterogeneity is a distinguishing feature of TLSs, which may lead to differences in their functions. Growing evidence suggests that TLSs are associated with various diseases, such as cancers, autoimmune diseases, transplant rejection, chronic inflammation, infection, and even ageing. However, the detailed mechanisms behind these clinical associations are not yet fully understood. The mechanisms by which TLS maturation and localization affect immune function are also unclear. Therefore, it is necessary to enhance the understanding of TLS development and function at the cellular and molecular level, which may allow us to utilize them to improve the immune microenvironment. In this review, we delve into the composition, formation mechanism, associations with diseases, and potential therapeutic applications of TLSs. Furthermore, we discuss the therapeutic implications of TLSs, such as their role as markers of therapeutic response and prognosis. Finally, we summarize various methods for detecting and targeting TLSs. Overall, we provide a comprehensive understanding of TLSs and aim to develop more effective therapeutic strategies.
Collapse
Affiliation(s)
- Lianyu Zhao
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Song Jin
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyao Wang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Zhe Zhang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Xuan Wang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Zhanwei Chen
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Xiaohui Wang
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Dongsheng Zhang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Haiwei Wu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| |
Collapse
|
3
|
Magliozzi R, Howell OW, Calabrese M, Reynolds R. Meningeal inflammation as a driver of cortical grey matter pathology and clinical progression in multiple sclerosis. Nat Rev Neurol 2023:10.1038/s41582-023-00838-7. [PMID: 37400550 DOI: 10.1038/s41582-023-00838-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/05/2023]
Abstract
Growing evidence from cerebrospinal fluid samples and post-mortem brain tissue from individuals with multiple sclerosis (MS) and rodent models indicates that the meninges have a key role in the inflammatory and neurodegenerative mechanisms underlying progressive MS pathology. The subarachnoid space and associated perivascular spaces between the membranes of the meninges are the access points for entry of lymphocytes, monocytes and macrophages into the brain parenchyma, and the main route for diffusion of inflammatory and cytotoxic molecules from the cerebrospinal fluid into the brain tissue. In addition, the meningeal spaces act as an exit route for CNS-derived antigens, immune cells and metabolites. A number of studies have demonstrated an association between chronic meningeal inflammation and a more severe clinical course of MS, suggesting that the build-up of immune cell aggregates in the meninges represents a rational target for therapeutic intervention. Therefore, understanding the precise cell and molecular mechanisms, timing and anatomical features involved in the compartmentalization of inflammation within the meningeal spaces in MS is vital. Here, we present a detailed review and discussion of the cellular, molecular and radiological evidence for a role of meningeal inflammation in MS, alongside the clinical and therapeutic implications.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy.
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Owain W Howell
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
- Institute of Life Sciences, Swansea University, Swansea, UK
| | - Massimiliano Calabrese
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Richard Reynolds
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
4
|
Biased IL-2 signals induce Foxp3-rich pulmonary lymphoid structures and facilitate long-term lung allograft acceptance in mice. Nat Commun 2023; 14:1383. [PMID: 36914624 PMCID: PMC10011523 DOI: 10.1038/s41467-023-36924-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Transplantation of solid organs can be life-saving in patients with end-stage organ failure, however, graft rejection remains a major challenge. In this study, by pre-conditioning with interleukin-2 (IL-2)/anti-IL-2 antibody complex treatment biased toward IL-2 receptor α, we achieved acceptance of fully mismatched orthotopic lung allografts that remained morphologically and functionally intact for more than 90 days in immunocompetent mice. These allografts are tolerated by the actions of forkhead box p3 (Foxp3)+ regulatory T (Treg) cells that home to the lung allografts. Although counts of circulating Treg cells rapidly return to baseline following cessation of IL-2 treatment, Foxp3+ Treg cells persist in peribronchial and peribronchiolar areas of the grafted lungs, forming organized clusters reminiscent of inducible tertiary lymphoid structures (iTLS). These iTLS in lung allografts are made of Foxp3+ Treg cells, conventional T cells, and B cells, as evidenced by using microscopy-based distribution and neighborhood analyses. Foxp3-transgenic mice with inducible and selective deletion of Foxp3+ cells are unable to form iTLS in lung allografts, and these mice acutely reject lung allografts. Collectively, we report that short-term, high-intensity and biased IL-2 pre-conditioning facilitates acceptance of vascularized and ventilated lung allografts without the need of immunosuppression, by inducing Foxp3-controlled iTLS formation within allografts.
Collapse
|
5
|
Michel E, Galen Hartwig M, Sommer W. Lung Retransplantation. Thorac Surg Clin 2022; 32:259-268. [PMID: 35512943 DOI: 10.1016/j.thorsurg.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Lung retransplantation remains the standard treatment of irreversible lung allograft failure. The most common indications for lung retransplantation are acute graft failure, chronic lung allograft dysfunction, and postoperative airway complications. Careful patient selection with regards to indications, anatomy, extrapulmonary organ dysfunction (specifically renal dysfunction), and immunologic consideration are of utmost importance. The conduct of the lung retransplantation operation is arduous with special considerations given to operative approach, type of surgery (single vs bilateral), use of extracorporeal circulatory support, and hematological management. Outcomes have improved significantly for most patients, nearing short and midterm outcomes of primary lung recipients in select cases.
Collapse
Affiliation(s)
- Eriberto Michel
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, 55 Fruit Street, Cox 630, Boston, MA 02114, USA
| | - Matthew Galen Hartwig
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University School of Medicine, DUMC 3863, Durham, NC 27710, USA.
| | - Wiebke Sommer
- Department of Cardiac Surgery, University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| |
Collapse
|
6
|
Zhang QY, Ye XP, Zhou Z, Zhu CF, Li R, Fang Y, Zhang RJ, Li L, Liu W, Wang Z, Song SY, Lu SY, Zhao SX, Lin JN, Song HD. Lymphocyte infiltration and thyrocyte destruction are driven by stromal and immune cell components in Hashimoto's thyroiditis. Nat Commun 2022; 13:775. [PMID: 35140214 PMCID: PMC8828859 DOI: 10.1038/s41467-022-28120-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 01/07/2022] [Indexed: 11/09/2022] Open
Abstract
Hashimoto's thyroiditis (HT) is the most common autoimmune disease characterized by lymphocytic infiltration and thyrocyte destruction. Dissection of the interaction between the thyroidal stromal microenvironment and the infiltrating immune cells might lead to a better understanding of HT pathogenesis. Here we show, using single-cell RNA-sequencing, that three thyroidal stromal cell subsets, ACKR1+ endothelial cells and CCL21+ myofibroblasts and CCL21+ fibroblasts, contribute to the thyroidal tissue microenvironment in HT. These cell types occupy distinct histological locations within the thyroid gland. Our experiments suggest that they might facilitate lymphocyte trafficking from the blood to thyroid tissues, and T cell zone CCL21+ fibroblasts may also promote the formation of tertiary lymphoid organs characteristic to HT. Our study also demonstrates the presence of inflammatory macrophages and dendritic cells expressing high levels of IL-1β in the thyroid, which may contribute to thyrocyte destruction in HT patients. Our findings thus provide a deeper insight into the cellular interactions that might prompt the pathogenesis of HT.
Collapse
Affiliation(s)
- Qian-Yue Zhang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiao-Ping Ye
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Zheng Zhou
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Department of geriatric endocrinology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chen-Fang Zhu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Discipline Construction Research Center of China Hospital Development Institute, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Rui Li
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ya Fang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Rui-Jia Zhang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Lu Li
- Department of Endocrinology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wei Liu
- Department of Endocrinology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zheng Wang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shi-Yang Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Sang-Yu Lu
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shuang-Xia Zhao
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jian-Nan Lin
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Huai-Dong Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Department of Endocrinology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
7
|
Bos S, Filby AJ, Vos R, Fisher AJ. Effector immune cells in Chronic Lung Allograft Dysfunction: a Systematic Review. Immunology 2022; 166:17-37. [PMID: 35137398 PMCID: PMC9426626 DOI: 10.1111/imm.13458] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/13/2022] [Accepted: 02/02/2022] [Indexed: 11/29/2022] Open
Abstract
Chronic lung allograft dysfunction (CLAD) remains the major barrier to long‐term survival after lung transplantation and improved insight into its underlying immunological mechanisms is critical to better understand the disease and to identify treatment targets. We systematically searched the electronic databases of PubMed and EMBASE for original research publications, published between January 2000 and April 2021, to comprehensively assess current evidence on effector immune cells in lung tissue and bronchoalveolar lavage fluid from lung transplant recipients with CLAD. Literature search revealed 1351 articles, 76 of which met the criteria for inclusion in our analysis. Our results illustrate significant complexity in both innate and adaptive immune cell responses in CLAD, along with presence of numerous immune cell products, including cytokines, chemokines and proteases associated with tissue remodelling. A clear link between neutrophils and eosinophils and CLAD incidence has been seen, in which eosinophils more specifically predisposed to restrictive allograft syndrome. The presence of cytotoxic and T‐helper cells in CLAD pathogenesis is well‐documented, although it is challenging to draw conclusions about their role in tissue processes from predominantly bronchoalveolar lavage data. In restrictive allograft syndrome, a more prominent humoral immune involvement with increased B cells, immunoglobulins and complement deposition is seen. Our evaluation of published studies over the last 20 years summarizes the complex multifactorial immunopathology of CLAD onset and progression. It highlights the phenotype of several key effector immune cells involved in CLAD pathogenesis, as well as the paucity of single cell resolution spatial studies in lung tissue from patients with CLAD.
Collapse
Affiliation(s)
- Saskia Bos
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, United Kingdom.,Institute of Transplantation, The Newcastle Upon Tyne Hospital NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Andrew J Filby
- Flow Cytometry Core and Innovation, Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Robin Vos
- Department of CHROMETA, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium.,University Hospitals Leuven, Dept. of Respiratory Diseases, Leuven, Belgium
| | - Andrew J Fisher
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, United Kingdom.,Institute of Transplantation, The Newcastle Upon Tyne Hospital NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
8
|
Suzuki Y, Oishi H, Kanehira M, Matsuda Y, Hirama T, Noda M, Okada Y. Effect of CTLA4-Ig on Obliterative Bronchiolitis in a Mouse Intrapulmonary Tracheal Transplantation Model. Ann Thorac Cardiovasc Surg 2021; 27:355-365. [PMID: 33980752 PMCID: PMC8684841 DOI: 10.5761/atcs.oa.20-00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objectives: One of the serious problems after lung transplantation is chronic lung allograft dysfunction (CLAD). Most CLAD patients pathologically characterized by obliterative bronchiolitis (OB). Cytotoxic T-lymphocyte-associated antigen 4 (CTLA4)-Ig is a combination protein of the Fc fragment of human IgG1 linked to the extracellular domain of CTLA4. The aim of the study was to examine the effect of CTLA4-Ig therapy on OB using a mouse intrapulmonary tracheal transplantation (IPTT) model. Methods: IPTT was performed between BALB/c (donor) and C57BL/6 (recipient) mice. Abatacept, which is a commercially available form of CTLA4-Ig, was intraperitoneally injected in recipient mice immediately after surgery, on days 7, 14, and 21. The mice in the control group received human IgG. Results: We performed semi-quantitative analysis of graft luminal obliteration at post-transplant day 28. We calculated the obliteration ratio of the lumen of the transplanted trachea in each case. The obliteration ratio was significantly lower in the CTLA4-Ig group than that in the control group (91.2 ± 2.1% vs. 47.8 ± 7.9%, p = 0.0008). Immunofluorescent staining revealed significantly decreased lymphoid neogenesis in the lung. Conclusions: CTLA4-Ig therapy attenuated tracheal obliteration with fibrous tissue in the mouse IPTT model. The attenuation of fibrous obliteration was correlated with the inhibition of lymphoid neogenesis.
Collapse
Affiliation(s)
- Yamato Suzuki
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Hisashi Oishi
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Masahiko Kanehira
- Center for Life Science Research, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Yasushi Matsuda
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan.,Department of Thoracic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Takashi Hirama
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Masafumi Noda
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
9
|
Pezzuto F, Lunardi F, Vadori M, Zampieri D, Casiraghi F, Azzollini N, Vuljan SE, Mammana M, Vedovelli L, Schiavon M, Gregori D, Cozzi E, Rea F, Calabrese F. Chronic lung allograft pathology lesions in two rat strain combinations. J Thorac Dis 2021; 13:2833-2843. [PMID: 34164175 PMCID: PMC8182524 DOI: 10.21037/jtd-20-3415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Chronic lung allograft dysfunction remains an obstacle to long-term survival after lung transplantation. Two phenotypes have been described: obliterative bronchiolitis and restrictive allograft syndrome. Preclinical models are essential to analyze chronic lung allograft dysfunction pathophysiology. Methods Orthotopic lung transplants from 38 Lewis into Fischer 344 (Lew→F344) and 67 Brown-Norway into Lewis (BN→Lew) rats were performed in our center in the last decade. We carefully reviewed and quantified all grafts with chronic rejection (40 cases) (18 Lew→F344, 22 BN→Lew) with the aim to investigate if histological changes of chronic lung allograft dysfunction could be also detected in rat grafts. Results All animals showed human reminiscent histological lesions. Early chronic rejection lesions were detected in BN→Lew. End-stage chronic rejection with features of obliterative bronchiolitis was observed in 33% of Lew→F344; end-stage with restrictive allograft syndrome chronic rejection in 67% and 80% of Lew→F344 and BN→Lew, respectively. BN→Lew showed higher grades of endotheliitis, vascular fibrosis, and lower grades of lymphoid aggregates than Lew→F344 (P=0.007, P=0.043, P=0.004, respectively). Conclusions Chronic rejection lesions in rat lung allografts mimic those in humans. The frequent occurrence of restrictive allograft syndrome-like lesions in BN→Lew may be related to a higher degree of mismatch in this strain combination. These animal models could allow future mechanistic studies to better understand chronic lung allograft dysfunction pathogenesis.
Collapse
Affiliation(s)
- Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | | | - Davide Zampieri
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | | | - Nadia Azzollini
- Mario Negri Institute for Pharmacological Research, Bergamo, Italy
| | - Stefania Edith Vuljan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Marco Mammana
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Luca Vedovelli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Marco Schiavon
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Dario Gregori
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Emanuele Cozzi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| |
Collapse
|
10
|
Yoshiyasu N, Sato M. Chronic lung allograft dysfunction post-lung transplantation: The era of bronchiolitis obliterans syndrome and restrictive allograft syndrome. World J Transplant 2020; 10:104-116. [PMID: 32864356 PMCID: PMC7428788 DOI: 10.5500/wjt.v10.i5.104] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 02/05/2023] Open
Abstract
Chronic lung allograft dysfunction (CLAD) following lung transplantation limits long-term survival considerably. The main reason for this is a lack of knowledge regarding the pathological condition and the establishment of treatment. The consensus statement from the International Society for Heart and Lung Transplantation on CLAD in 2019 classified CLAD into two main phenotypes: Bronchiolitis obliterans syndrome and restrictive allograft syndrome. Along with this clear classification, further exploration of the mechanisms and the development of appropriate prevention and treatment strategies for each phenotype are desired. In this review, we summarize the new definition of CLAD and update and summarize the existing knowledge on the underlying mechanisms of bronchiolitis obliterans syndrome and restrictive allograft syndrome, which have been elucidated from clinicopathological observations and animal experiments worldwide.
Collapse
Affiliation(s)
- Nobuyuki Yoshiyasu
- Department of Thoracic Surgery, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Masaaki Sato
- Department of Thoracic Surgery, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| |
Collapse
|
11
|
Sato M. Bronchiolitis obliterans syndrome and restrictive allograft syndrome after lung transplantation: why are there two distinct forms of chronic lung allograft dysfunction? ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:418. [PMID: 32355862 PMCID: PMC7186721 DOI: 10.21037/atm.2020.02.159] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bronchiolitis obliterans syndrome (BOS) had been considered to be the representative form of chronic rejection or chronic lung allograft dysfunction (CLAD) after lung transplantation. In BOS, small airways are affected by chronic inflammation and obliterative fibrosis, whereas peripheral lung tissue remains relatively intact. However, recognition of another form of CLAD involving multiple tissue compartments in the lung, termed restrictive allograft syndrome (RAS), raised a fundamental question: why there are two phenotypes of CLAD? Increasing clinical and experimental data suggest that RAS may be a prototype of chronic rejection after lung transplantation involving both cellular and antibody-mediated alloimmune responses. Some cases of RAS are also induced by fulminant general inflammation in lung allografts. However, BOS involves alloimmune responses and the airway-centered disease process can be explained by multiple mechanisms such as external alloimmune-independent stimuli (such as infection, aspiration and air pollution), exposure of airway-specific autoantigens and airway ischemia. Localization of immune responses in different anatomical compartments in different phenotypes of CLAD might be associated with lymphoid neogenesis or the de novo formation of lymphoid tissue in lung allografts. Better understanding of distinct mechanisms of BOS and RAS will facilitate the development of effective preventive and therapeutic strategies of CLAD.
Collapse
Affiliation(s)
- Masaaki Sato
- Department of Thoracic Surgery, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
12
|
Blokland SLM, Flessa CM, van Roon JAG, Mavragani CP. Emerging roles for chemokines and cytokines as orchestrators of immunopathology in Sjögren's syndrome. Rheumatology (Oxford) 2019; 60:3072-3087. [PMID: 30838419 DOI: 10.1093/rheumatology/key438] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/03/2018] [Indexed: 12/14/2022] Open
Abstract
In primary SS (pSS), chemokines and cytokines orchestrate immunopathology driven by a complex network of interacting inflammatory cells. In recent years, the importance of chemotactic and non-chemotactic cytokines that control function, movement and placing of all cells within the inflamed exocrine glands and directing immunopathology has become increasingly clear. This paper reviews the current knowledge on chemokines and focuses on the emerging roles of novel chemotactic and non-chemotactic mediators in pSS. It highlights their contribution to pathogenic processes such as B cell hyperactivity and the formation of ectopic lymphoid structures. To this end, the role of acquired (CXCR5/CCR9 Th-cell-mediated) and innate (inflammasome/IL-1/IL-18-mediated) pathways in steering immunopathology is discussed.
Collapse
Affiliation(s)
- Sofie L M Blokland
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, The Netherlands.,Laboratory of Translational Immunology, Department of Immunology, Utrecht University, Utrecht, The Netherlands
| | - Christina-Maria Flessa
- Department of Physiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Joel A G van Roon
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, The Netherlands.,Laboratory of Translational Immunology, Department of Immunology, Utrecht University, Utrecht, The Netherlands
| | - Clio P Mavragani
- Department of Physiology, National and Kapodistrian University of Athens, Athens, Greece.,Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
13
|
Abstract
Pulmonary respiration inevitably exposes the mucosal surface of the lung to potentially noxious stimuli, including pathogens, allergens, and particulates, each of which can trigger pulmonary damage and inflammation. As inflammation resolves, B and T lymphocytes often aggregate around large bronchi to form inducible Bronchus-Associated Lymphoid Tissue (iBALT). iBALT formation can be initiated by a diverse array of molecular pathways that converge on the activation and differentiation of chemokine-expressing stromal cells that serve as the scaffolding for iBALT and facilitate the recruitment, retention, and organization of leukocytes. Like conventional lymphoid organs, iBALT recruits naïve lymphocytes from the blood, exposes them to local antigens, in this case from the airways, and supports their activation and differentiation into effector cells. The activity of iBALT is demonstrably beneficial for the clearance of respiratory pathogens; however, it is less clear whether it dampens or exacerbates inflammatory responses to non-infectious agents. Here, we review the evidence regarding the role of iBALT in pulmonary immunity and propose that the final outcome depends on the context of the disease.
Collapse
|
14
|
Jansen CS, Prokhnevska N, Kissick HT. The requirement for immune infiltration and organization in the tumor microenvironment for successful immunotherapy in prostate cancer. Urol Oncol 2018; 37:543-555. [PMID: 30446449 DOI: 10.1016/j.urolonc.2018.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/01/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022]
Abstract
Immunotherapy-particularly immune checkpoint blockade-has seen great success in many tumor types. However, checkpoint-based therapies have not demonstrated high levels of success in prostate cancer, and there is much to be learned from both the successes and failures of these treatments. Here we review the evidence that composition of infiltrating immune cells in the tumor microenvironment is fundamental to the response to immunotherapy. Additionally, we discuss the emerging idea that the organization of these immune cells may also be crucial to this response. In prostate cancer, the composition and organization of the tumor immune microenvironment are preeminent topics of discussion and areas of important future investigation.
Collapse
Affiliation(s)
| | | | - Haydn T Kissick
- Department of Urology, Emory University, Atlanta, GA; Department of Microbiology and Immunology, Emory University, Atlanta, GA.
| |
Collapse
|
15
|
Yamamoto H, Sugimoto S, Tanaka S, Kurosaki T, Otani S, Yamane M, Taira N, Oto T, Toyooka S. A single-nucleotide polymorphism in a gene modulating glucocorticoid sensitivity is associated with the decline in total lung capacity after lung transplantation. Surg Today 2018; 49:268-274. [DOI: 10.1007/s00595-018-1717-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/09/2018] [Indexed: 01/14/2023]
|
16
|
Pipi E, Nayar S, Gardner DH, Colafrancesco S, Smith C, Barone F. Tertiary Lymphoid Structures: Autoimmunity Goes Local. Front Immunol 2018; 9:1952. [PMID: 30258435 PMCID: PMC6143705 DOI: 10.3389/fimmu.2018.01952] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are frequently observed in target organs of autoimmune diseases. TLS present features of secondary lymphoid organs such as segregated T and B cell zones, presence of follicular dendritic cell networks, high endothelial venules and specialized lymphoid fibroblasts and display the mechanisms to support local adaptive immune responses toward locally displayed antigens. TLS detection in the tissue is often associated with poor prognosis of disease, auto-antibody production and malignancy development. This review focuses on the contribution of TLS toward the persistence of the inflammatory drive, the survival of autoreactive lymphocyte clones and post-translational modifications, responsible for the pathogenicity of locally formed autoantibodies, during autoimmune disease development.
Collapse
Affiliation(s)
- Elena Pipi
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,Experimental Medicine Unit, Immuno-Inflammation Therapeutic Area, GSK Medicines Research Centre, Stevenage, United Kingdom
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - David H Gardner
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | | | - Charlotte Smith
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
17
|
Ledderose C, Liu K, Kondo Y, Slubowski CJ, Dertnig T, Denicoló S, Arbab M, Hubner J, Konrad K, Fakhari M, Lederer JA, Robson SC, Visner GA, Junger WG. Purinergic P2X4 receptors and mitochondrial ATP production regulate T cell migration. J Clin Invest 2018; 128:3583-3594. [PMID: 29894310 DOI: 10.1172/jci120972] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
T cells must migrate in order to encounter antigen-presenting cells (APCs) and to execute their varied functions in immune defense and inflammation. ATP release and autocrine signaling through purinergic receptors contribute to T cell activation at the immune synapse that T cells form with APCs. Here, we show that T cells also require ATP release and purinergic signaling for their migration to APCs. We found that the chemokine stromal-derived factor-1α (SDF-1α) triggered mitochondrial ATP production, rapid bursts of ATP release, and increased migration of primary human CD4+ T cells. This process depended on pannexin-1 ATP release channels and autocrine stimulation of P2X4 receptors. SDF-1α stimulation caused localized accumulation of mitochondria with P2X4 receptors near the front of cells, resulting in a feed-forward signaling mechanism that promotes cellular Ca2+ influx and sustains mitochondrial ATP synthesis at levels needed for pseudopod protrusion, T cell polarization, and cell migration. Inhibition of P2X4 receptors blocked the activation and migration of T cells in vitro. In a mouse lung transplant model, P2X4 receptor antagonist treatment prevented the recruitment of T cells into allograft tissue and the rejection of lung transplants. Our findings suggest that P2X4 receptors are therapeutic targets for immunomodulation in transplantation and inflammatory diseases.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kaifeng Liu
- Department of Medicine/Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yutaka Kondo
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian J Slubowski
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Dertnig
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara Denicoló
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mona Arbab
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Johannes Hubner
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kirstin Konrad
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mahtab Fakhari
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Simon C Robson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Gary A Visner
- Department of Medicine/Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Ludwig Boltzmann Institute for Traumatology, Vienna, Austria
| |
Collapse
|
18
|
Nerviani A, Pitzalis C. Role of chemokines in ectopic lymphoid structures formation in autoimmunity and cancer. J Leukoc Biol 2018; 104:333-341. [PMID: 29947426 PMCID: PMC6099300 DOI: 10.1002/jlb.3mr0218-062r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/09/2018] [Accepted: 05/21/2018] [Indexed: 12/19/2022] Open
Abstract
Ectopic (or tertiary) lymphoid structures (ELS) are organized aggregates of lymphocytes resembling secondary lymphoid organs and developing in chronically inflamed nonlymphoid tissues during persistent infections, graft rejection, autoimmune conditions, and cancer. In this review, we will first depict the mechanisms regulating ELS generation, focusing on the role played by lymphoid chemokines. We will then characterize ELS forming in target organs during autoimmune conditions, here exemplified by rheumatoid arthritis, and cancer, highlighting the relevance of the tissue-specific factors. Finally, we will discuss the clinical significance of ELS and the therapeutic potential of their inhibition and/or enhancement depending on the disease considered.
Collapse
Affiliation(s)
- Alessandra Nerviani
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
19
|
Pentraxin 3 deficiency enhances features of chronic rejection in a mouse orthotopic lung transplantation model. Oncotarget 2018; 9:8489-8501. [PMID: 29492210 PMCID: PMC5823599 DOI: 10.18632/oncotarget.23902] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/12/2017] [Indexed: 12/24/2022] Open
Abstract
Chronic lung allograft dysfunction (CLAD) is a serious complication after lung transplantation and thought to represent chronic rejection. Increased expression of Pentraxin 3 (PTX3), an acute phase protein, was associated with worse outcome in lung transplant patients. To determine the role of recipient PTX3 in development of chronic rejection, we used a minor alloantigen-mismatched murine orthotopic single lung transplant model. Male C57BL/10 mice were used as donors. Male PTX3 knockout (KO) mice and their wild type (WT) littermates on 129/SvEv/C57BL6/J background were used as recipients. In KO recipients, 7/13 grafted lungs were consolidated without volume recovery on CT scan, while only 2/9 WT mice showed similar graft consolidation. For grafts where lung volume could be reliably analyzed by CT scan, the lung volume recovery was significantly reduced in KO mice compared to WT. Interstitial inflammation, parenchymal fibrosis and bronchiolitis obliterans scores were significantly higher in KO mice. Presence of myofibroblasts and lymphoid aggregation was significantly enhanced in the grafts of PTX3 KO recipients. Recipient PTX3 deficiency enhanced chronic rejection-like lesions by promoting a fibrotic process in the airways and lung parenchyma. The underlying mechanisms and potential protective role of exogenous PTX3 as a therapy should be further explored.
Collapse
|
20
|
Mueller CG, Nayar S, Campos J, Barone F. Molecular and Cellular Requirements for the Assembly of Tertiary Lymphoid Structures. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:55-72. [PMID: 30155622 DOI: 10.1007/978-3-319-78127-3_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
At sites of chronic inflammation, recruited immune cells form structures that resemble secondary lymphoid organs (SLOs). Those are characterized by segregated areas of prevalent T- or B-cell aggregation, differentiation of high endothelial venules (HEVs) and local activation of resident stromal cells. B-cell proliferation and affinity maturation towards locally displayed autoantigens have been demonstrated at those sites, known as tertiary lymphoid structures (TLSs). TLS formation has been associated with local disease persistence and progression as well as increased systemic manifestations. While bearing a similar histological structure to SLO, the signals that regulate TLS and SLO formation can diverge, and a series of pro-inflammatory cytokines has been ascribed as responsible for TLS formation at different anatomical sites. Here we review the structural elements as well as the signals responsible for TLS aggregation, aiming to provide an overview to this complex immunological phenomenon.
Collapse
Affiliation(s)
- C G Mueller
- CNRS UPR 3572, Laboratory of Immunopathology and Therapeutic Chemistry/Laboratory of Excellence MEDALIS, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - S Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham, UK
| | - J Campos
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham, UK
| | - F Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham, UK.
| |
Collapse
|
21
|
Alsughayyir J, Pettigrew GJ, Motallebzadeh R. Spoiling for a Fight: B Lymphocytes As Initiator and Effector Populations within Tertiary Lymphoid Organs in Autoimmunity and Transplantation. Front Immunol 2017; 8:1639. [PMID: 29218052 PMCID: PMC5703719 DOI: 10.3389/fimmu.2017.01639] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 11/09/2017] [Indexed: 12/13/2022] Open
Abstract
Tertiary lymphoid organs (TLOs) develop at ectopic sites within chronically inflamed tissues, such as in autoimmunity and rejecting organ allografts. TLOs differ structurally from canonical secondary lymphoid organs (SLOs), in that they lack a mantle zone and are not encapsulated, suggesting that they may provide unique immune function. A notable feature of TLOs is the frequent presence of structures typical of germinal centers (GCs). However, little is known about the role of such GCs, and in particular, it is not clear if the B cell response within is autonomous, or whether it synergizes with concurrent responses in SLOs. This review will discuss ectopic lymphoneogenesis and the role of the B cell in TLO formation and subsequent effector output in the context of autoimmunity and transplantation, with particular focus on the contribution of ectopic GCs to affinity maturation in humoral immune responses and to the potential breakdown of self-tolerance and development of humoral autoimmunity.
Collapse
Affiliation(s)
- Jawaher Alsughayyir
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Gavin J Pettigrew
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Reza Motallebzadeh
- Division of Surgery and Interventional Science, University College London, London, United Kingdom.,Institute of Immunity and Transplantation, University College London, London, United Kingdom.,Department of Nephrology, Urology and Transplantation, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
22
|
Weigt SS, Wang X, Palchevskiy V, Gregson AL, Patel N, DerHovanessian A, Shino MY, Sayah DM, Birjandi S, Lynch JP, Saggar R, Ardehali A, Ross DJ, Palmer SM, Elashoff D, Belperio JA. Gene Expression Profiling of Bronchoalveolar Lavage Cells Preceding a Clinical Diagnosis of Chronic Lung Allograft Dysfunction. PLoS One 2017; 12:e0169894. [PMID: 28103284 PMCID: PMC5245825 DOI: 10.1371/journal.pone.0169894] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 12/22/2016] [Indexed: 11/18/2022] Open
Abstract
Background Chronic Lung Allograft Dysfunction (CLAD) is the main limitation to long-term survival after lung transplantation. Although CLAD is usually not responsive to treatment, earlier identification may improve treatment prospects. Methods In a nested case control study, 1-year post transplant surveillance bronchoalveolar lavage (BAL) fluid samples were obtained from incipient CLAD (n = 9) and CLAD free (n = 8) lung transplant recipients. Incipient CLAD cases were diagnosed with CLAD within 2 years, while controls were free from CLAD for at least 4 years following bronchoscopy. Transcription profiles in the BAL cell pellets were assayed with the HG-U133 Plus 2.0 microarray (Affymetrix). Differential gene expression analysis, based on an absolute fold change (incipient CLAD vs no CLAD) >2.0 and an unadjusted p-value ≤0.05, generated a candidate list containing 55 differentially expressed probe sets (51 up-regulated, 4 down-regulated). Results The cell pellets in incipient CLAD cases were skewed toward immune response pathways, dominated by genes related to recruitment, retention, activation and proliferation of cytotoxic lymphocytes (CD8+ T-cells and natural killer cells). Both hierarchical clustering and a supervised machine learning tool were able to correctly categorize most samples (82.3% and 94.1% respectively) into incipient CLAD and CLAD-free categories. Conclusions These findings suggest that a pathobiology, similar to AR, precedes a clinical diagnosis of CLAD. A larger prospective investigation of the BAL cell pellet transcriptome as a biomarker for CLAD risk stratification is warranted.
Collapse
Affiliation(s)
- S. Samuel Weigt
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- * E-mail:
| | - Xiaoyan Wang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Vyacheslav Palchevskiy
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Aric L. Gregson
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Naman Patel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Ariss DerHovanessian
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Michael Y. Shino
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - David M. Sayah
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Shirin Birjandi
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Joseph P. Lynch
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Rajan Saggar
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Abbas Ardehali
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - David J. Ross
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Scott M. Palmer
- Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - David Elashoff
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - John A. Belperio
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| |
Collapse
|
23
|
Neyt K, GeurtsvanKessel CH, Deswarte K, Hammad H, Lambrecht BN. Early IL-1 Signaling Promotes iBALT Induction after Influenza Virus Infection. Front Immunol 2016; 7:312. [PMID: 27579026 PMCID: PMC4985557 DOI: 10.3389/fimmu.2016.00312] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/02/2016] [Indexed: 11/13/2022] Open
Abstract
Inducible bronchus-associated lymphoid tissue (iBALT) is a long lasting tertiary lymphoid tissue that can be induced following influenza A virus (IAV) infection. Previous studies have shown that iBALT structures containing germinal center (GC) B cells protect against repeated infection by contributing locally to the cellular and humoral immune response. If we are to exploit this in vaccination strategies, we need a better understanding on how iBALT structures are induced. One hypothesis is that the strength of the initial innate response dictates induction of iBALT. In the present study, we investigated the role of interleukin (IL)-1 and IL-1R signaling on iBALT formation. Mice lacking the IL-1R had a delayed viral clearance and, thus, a prolonged exposure to viral replication, leading to increased disease severity, compared to wild-type mice. Contradictorily, iBALT formation following clearance of the virus was heavily compromised in Il1r1−/− mice. Quantification of gene induction after IAV infection demonstrated induction of IL-1α and to a much lesser extent of IL-1β. Administration of recombinant IL-1α to the lungs of wild-type mice, early but not late, after IAV infection led to more pronounced iBALT formation and an increased amount of GC B cells in the lungs. Bone marrow chimeric mice identified the stromal compartment as the crucial IL-1 responsive cell for iBALT induction. Mechanistically, Q-PCR analysis of lung homogenates revealed a strongly diminished production of CXCL13, a B cell-attracting chemokine, in Il1r−/− mice during the early innate phase of IAV infection. These experiments demonstrate that appropriate innate IL-1α–IL-1R signaling is necessary for IAV clearance and at the same time instructs the formation of organized tertiary lymphoid tissues through induction of CXCL13 early after infection. These findings are discussed in the light of recent insights on the pathogenesis of tertiary lymphoid organ formation in the lung in various diseases where the IL-1 axis is hyperactive, such as rheumatoid arthritis and COPD.
Collapse
Affiliation(s)
- Katrijn Neyt
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | | | - Kim Deswarte
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
24
|
Kranich J, Krautler NJ. How Follicular Dendritic Cells Shape the B-Cell Antigenome. Front Immunol 2016; 7:225. [PMID: 27446069 PMCID: PMC4914831 DOI: 10.3389/fimmu.2016.00225] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 05/26/2016] [Indexed: 12/21/2022] Open
Abstract
Follicular dendritic cells (FDCs) are stromal cells residing in primary follicles and in germinal centers of secondary and tertiary lymphoid organs (SLOs and TLOs). There, they play a crucial role in B-cell activation and affinity maturation of antibodies. FDCs have the unique capacity to bind and retain native antigen in B-cell follicles for long periods of time. Therefore, FDCs shape the B-cell antigenome (the sum of all B-cell antigens) in SLOs and TLOs. In this review, we discuss recent findings that explain how this stromal cell type can arise in almost any tissue during TLO formation and, furthermore, focus on the mechanisms of antigen capture and retention involved in the generation of long-lasting antigen depots displayed on FDCs.
Collapse
Affiliation(s)
- Jan Kranich
- Institute for Immunology, Ludwig Maximilian University Munich, Munich, Germany
| | | |
Collapse
|
25
|
Matsuda Y, Wang X, Oishi H, Guan Z, Saito M, Liu M, Keshavjee S, Chow CW. Spleen Tyrosine Kinase Modulates Fibrous Airway Obliteration and Associated Lymphoid Neogenesis After Transplantation. Am J Transplant 2016; 16:342-52. [PMID: 26308240 DOI: 10.1111/ajt.13442] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 06/16/2015] [Accepted: 06/21/2015] [Indexed: 01/25/2023]
Abstract
Chronic lung allograft dysfunction, the major cause of death following lung transplantation, usually manifests as irreversible airflow obstruction associated with obliterative bronchiolitis (OB), a lesion characterized by chronic inflammation, lymphoid neogenesis, fibroproliferation and small airway obliteration. Spleen tyrosine kinase (Syk), a tyrosine kinase that regulates B cell function and innate immunity, has been implicated in the pathogenesis of chronic inflammation and tissue repair. This study evaluated the role of Syk in development of OB, using an intrapulmonary tracheal transplant model of OB with the conditional Syk-knockout Syk(flox/flox) //rosa26-CreER(T2) mice and a Syk-selective inhibitor, GSK2230413. BALB/c trachea allografts were transplanted into Syk-knockout (Syk(del/del) ) mice or wild-type C57BL/6 recipients treated with GSK2230413. At day 28, histological analysis revealed that in the Syk(del/del) and GSK2230413-treated C57BL/6 recipients, the graft lumen remained open compared with allografts transplanted into Syk-expressing (Syk(flox/flox) ) and placebo control-treated C57BL/6 recipients. Immunofluorescence showed lymphoid neogenesis with distinct B and T cell zones in control mice. In contrast, lymphoid neogenesis was absent and few B or T cells were found in Syk(del/del) and GSK2230413-treated mice. These observations suggest that inhibition of Syk may be a potential therapeutic strategy for the management of OB following lung transplantation.
Collapse
Affiliation(s)
- Y Matsuda
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - X Wang
- Division of Respirology, Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - H Oishi
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - Z Guan
- Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - M Saito
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - M Liu
- Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - S Keshavjee
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - C-W Chow
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Division of Respirology, Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
26
|
Jones GW, Jones SA. Ectopic lymphoid follicles: inducible centres for generating antigen-specific immune responses within tissues. Immunology 2015; 147:141-51. [PMID: 26551738 PMCID: PMC4717241 DOI: 10.1111/imm.12554] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 10/28/2015] [Accepted: 11/01/2015] [Indexed: 02/06/2023] Open
Abstract
Lymphoid neogenesis is traditionally viewed as a pre‐programmed process that promotes the formation of lymphoid organs during development. Here, the spatial organization of T and B cells in lymph nodes and spleen into discrete structures regulates antigen‐specific responses and adaptive immunity following immune challenge. However, lymphoid neogenesis is also triggered by chronic or persistent inflammation. Here, ectopic (or tertiary) lymphoid organs frequently develop in inflamed tissues as a response to infection, auto‐immunity, transplantation, cancer or environmental irritants. Although these structures affect local immune responses, the contribution of these lymphoid aggregates to the underlining pathology are highly context dependent and can elicit either protective or deleterious outcomes. Here we review the cellular and molecular mechanisms responsible for ectopic lymphoid neogenesis and consider the relevance of these structures in human disease.
Collapse
Affiliation(s)
- Gareth W Jones
- Division of Infection and Immunity, The School of Medicine, Cardiff University, Cardiff, UK
| | - Simon A Jones
- Division of Infection and Immunity, The School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
27
|
Wittwer T, Rahmanian P, Choi YH, Zeriouh M, Karavidic S, Neef K, Christmann A, Piatkowski T, Schnapper A, Ochs M, Mühlfeld C, Wahlers T. Mesenchymal stem cell pretreatment of non-heart-beating-donors in experimental lung transplantation. J Cardiothorac Surg 2014; 9:151. [PMID: 25179441 PMCID: PMC4169637 DOI: 10.1186/s13019-014-0151-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 08/18/2014] [Indexed: 12/30/2022] Open
Abstract
Background Lung transplantation (LTx) is still limited by organ shortage. To expand the donor pool, lung retrieval from non-heart-beating donors (NHBD) was introduced into clinical practice recently. However, primary graft dysfunction with inactivation of endogenous surfactant due to ischemia/reperfusion-injury is a major cause of early mortality. Furthermore, donor-derived human mesenchymal stem cell (hMSC) expansion and fibrotic differentiation in the allograft results in bronchiolitis obliterans syndrome (BOS), a leading cause of post-LTx long-term mortality. Therefore, pretreatment of NHBD with recipient-specific bone-marrow-(BM)-derived hMSC might have the potential to both improve the postischemic allograft function and influence the long-term development of BOS by the numerous paracrine, immunomodulating and tissue-remodeling properties especially on type-II-pneumocytes of hMSC. Methods Asystolic pigs (n = 5/group) were ventilated for 3 h of warm ischemia (groups 2–4). 50x106 mesenchymal-stem-cells (MSC) were administered in the pulmonary artery (group 3) or nebulized endobronchially (group 4) before lung preservation. Following left-lung-transplantation, grafts were reperfused, pulmonary-vascular-resistance (PVR), oxygenation and dynamic-lung-compliance (DLC) were monitored and compared to control-lungs (group 2) and sham-controls (group 1). To prove and localize hMSC in the lung, cryosections were counter-stained. Intra-alveolar edema was determined stereologically. Statistics comprised ANOVA with repeated measurements. Results Oxygenation (p = 0.001) and PVR (p = 0.009) following endovascular application of hMSC were significantly inferior compared to Sham controls, whereas DLC was significantly higher in endobronchially pretreated lungs (p = 0.045) with overall sham-comparable outcome regarding oxygenation and PVR. Stereology revealed low intrapulmonary edema in all groups (p > 0.05). In cryosections of both unreperfused and reperfused grafts, hMSC were localized in vessels of alveolar septa (endovascular application) and alveolar lumen (endobronchial application), respectively. Conclusions Preischemic deposition of hMSC in donor lungs is feasible and effective, and endobronchial application is associated with significantly better DLC as compared to sham controls. In contrast, transvascular hMSC delivery results in inferior oxygenation and PVR. In the long term perspective, due to immunomodulatory, paracrine and tissue-remodeling effects on epithelial and endothelial restitution, an endobronchial NHBD allograft-pretreatment with autologous mesenchymal-stem-cells to attenuate limiting bronchiolitis-obliterans-syndrome in the long-term perspective might be promising in clinical lung transplantation. Subsequent work with chronic experiments is initiated to further elucidate this important field. Electronic supplementary material The online version of this article (doi:10.1186/s13019-014-0151-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Thorsten Wittwer
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Kerpener Strasse 61, Cologne, 50924, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Akhavanpoor M, Wangler S, Gleissner CA, Korosoglou G, Katus HA, Erbel C. Adventitial inflammation and its interaction with intimal atherosclerotic lesions. Front Physiol 2014; 5:296. [PMID: 25152736 PMCID: PMC4126462 DOI: 10.3389/fphys.2014.00296] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 07/22/2014] [Indexed: 12/22/2022] Open
Abstract
The presence of adventitial inflammation in correlation with atherosclerotic lesions has been recognized for decades. In the last years, several studies have investigated the relevance and impact of adventitial inflammation on atherogenesis. In the abdominal aorta of elderly Apoe−/− mice, adventitial inflammatory structures were characterized as organized ectopic lymphoid tissue, and therefore termed adventitial tertiary lymphoid organs (ATLOs). These ATLOs possess similarities in development, structure and function to secondary lymphoid organs. A crosstalk between intimal atherosclerotic lesions and ATLOs has been suggested, and several studies could demonstrate a potential role for medial vascular smooth muscle cells in this process. We here review the development, phenotypic characteristics, and function of ATLOs in atherosclerosis. Furthermore, we discuss the possible role of medial vascular smooth muscle cells and their interaction between plaque and ATLOs.
Collapse
Affiliation(s)
- Mohammadreza Akhavanpoor
- Department of Cardiology, University of Heidelberg Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany
| | - Susanne Wangler
- Department of Cardiology, University of Heidelberg Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany
| | - Christian A Gleissner
- Department of Cardiology, University of Heidelberg Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany
| | - Grigorios Korosoglou
- Department of Cardiology, University of Heidelberg Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany
| | - Hugo A Katus
- Department of Cardiology, University of Heidelberg Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany
| | - Christian Erbel
- Department of Cardiology, University of Heidelberg Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany
| |
Collapse
|
29
|
Pitzalis C, Jones GW, Bombardieri M, Jones SA. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat Rev Immunol 2014; 14:447-62. [PMID: 24948366 DOI: 10.1038/nri3700] [Citation(s) in RCA: 520] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ectopic lymphoid-like structures often develop at sites of inflammation where they influence the course of infection, autoimmune disease, cancer and transplant rejection. These lymphoid aggregates range from tight clusters of B cells and T cells to highly organized structures that comprise functional germinal centres. Although the mechanisms governing ectopic lymphoid neogenesis in human pathology remain poorly defined, the presence of ectopic lymphoid-like structures within inflamed tissues has been linked to both protective and deleterious outcomes in patients. In this Review, we discuss investigations in both experimental model systems and patient cohorts to provide a perspective on the formation and functions of ectopic lymphoid-like structures in human pathology, with particular reference to the clinical implications and the potential for therapeutic targeting.
Collapse
Affiliation(s)
- Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Gareth W Jones
- Cardiff Institute for Infection and Immunity, The School of Medicine, Cardiff University, The Tenovus Building, Heath Campus, Cardiff CF14 4XN, Wales, UK
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Simon A Jones
- Cardiff Institute for Infection and Immunity, The School of Medicine, Cardiff University, The Tenovus Building, Heath Campus, Cardiff CF14 4XN, Wales, UK
| |
Collapse
|
30
|
Abstract
The enduring success of lung transplantation is built on the use of immunosuppressive drugs to stop the immune system from rejecting the newly transplanted lung allograft. Most patients receive a triple-drug maintenance immunosuppressive regimen consisting of a calcineurin inhibitor, an antiproliferative and corticosteroids. Induction therapy with either an antilymphocyte monoclonal or an interleukin-2 receptor antagonist are prescribed by many centres aiming to achieve rapid inhibition of recently activated and potentially alloreactive T lymphocytes. Despite this generic approach acute rejection episodes remain common, mandating further fine-tuning and augmentation of the immunosuppressive regimen. While there has been a trend away from cyclosporine and azathioprine towards a preference for tacrolimus and mycophenolate mofetil, this has not translated into significant protection from the development of chronic lung allograft dysfunction, the main barrier to the long-term success of lung transplantation. This article reviews the problem of lung allograft rejection and the evidence for immunosuppressive regimens used both in the short- and long-term in patients undergoing lung transplantation.
Collapse
|
31
|
Mohanta SK, Yin C, Peng L, Srikakulapu P, Bontha V, Hu D, Weih F, Weber C, Gerdes N, Habenicht AJ. Artery Tertiary Lymphoid Organs Contribute to Innate and Adaptive Immune Responses in Advanced Mouse Atherosclerosis. Circ Res 2014; 114:1772-87. [PMID: 24855201 DOI: 10.1161/circresaha.114.301137] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tertiary lymphoid organs emerge in tissues in response to nonresolving inflammation. Recent research characterized artery tertiary lymphoid organs in the aorta adventitia of aged apolipoprotein E–deficient mice. The atherosclerosis-associated lymphocyte aggregates are organized into distinct compartments, including separate T-cell areas harboring conventional, monocyte-derived, lymphoid, and plasmacytoid dendritic cells, as well as activated T-cell effectors and memory cells; B-cell follicles containing follicular dendritic cells in activated germinal centers; and peripheral niches of plasma cells. Artery tertiary lymphoid organs show marked neoangiogenesis, aberrant lymphangiogenesis, and extensive induction of high endothelial venules. Moreover, newly formed lymph node–like conduits connect the external lamina with high endothelial venules in T-cell areas and also extend into germinal centers. Mouse artery tertiary lymphoid organs recruit large numbers of naïve T cells and harbor lymphocyte subsets with opposing activities, including CD4
+
and CD8
+
effector and memory T cells, natural and induced CD4
+
regulatory T cells, and memory B cells at different stages of differentiation. These data suggest that artery tertiary lymphoid organs participate in primary immune responses and organize T- and B-cell autoimmune responses in advanced atherosclerosis. In this review, we discuss the novel concept that pro- and antiatherogenic immune responses toward unknown arterial wall–derived autoantigens may be organized by artery tertiary lymphoid organs and that disruption of the balance between pro- and antiatherogenic immune cell subsets may trigger clinically overt atherosclerosis.
Collapse
Affiliation(s)
- Sarajo Kumar Mohanta
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Changjun Yin
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Li Peng
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Prasad Srikakulapu
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Vineela Bontha
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Desheng Hu
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Falk Weih
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Christian Weber
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Norbert Gerdes
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Andreas J.R. Habenicht
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| |
Collapse
|
32
|
Saito T, Takahashi H, Kaneda H, Binnie M, Azad S, Sato M, Waddell TK, Cypel M, Liu M, Keshavjee S. Impact of cytokine expression in the pre-implanted donor lung on the development of chronic lung allograft dysfunction subtypes. Am J Transplant 2013; 13:3192-201. [PMID: 24164971 DOI: 10.1111/ajt.12492] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/13/2013] [Accepted: 09/07/2013] [Indexed: 01/25/2023]
Abstract
The long-term success of lung transplantation continues to be challenged by the development of chronic lung allograft dysfunction (CLAD). The purpose of this study was to investigate the relationship between cytokine expression levels in pre-implanted donor lungs and the posttransplant development of CLAD and its subtypes, bronchiolitis obliterans syndrome (BOS) and restrictive allograft syndrome (RAS). Of 109 patients who underwent bilateral lung or heart-lung transplantation and survived for more than 3 months, 50 BOS, 21 RAS and 38 patients with No CLAD were identified by pulmonary function test results. Using donor lung tissue biopsies sampled from each patient, expression levels of IL-6, IL-1β, IL-8, IL-10, interferon-γ and tumor necrosis factor-α mRNA were measured. IL-6 expression levels were significantly higher in pre-implanted lungs of patients that ultimately developed BOS compared to RAS and No CLAD (p = 0.025 and 0.011, respectively). Cox regression analysis demonstrated an association between high IL-6 expression levels and BOS development (hazard ratio = 4.98; 95% confidence interval = 2.42-10.2, p < 0.001). In conclusion, high IL-6 mRNA expression levels in pre-implanted donor lungs were associated with the development of BOS, not RAS. This association further supports the contention that early graft injury impacts on both late graft function and early graft function.
Collapse
Affiliation(s)
- T Saito
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Thoracic and Cardiovascular Surgery, Kansai Medical University, Hirakara, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen L, Fabian KL, Taylor JL, Storkus WJ. Therapeutic use of dendritic cells to promote the extranodal priming of anti-tumor immunity. Front Immunol 2013; 4:388. [PMID: 24348473 PMCID: PMC3843121 DOI: 10.3389/fimmu.2013.00388] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/05/2013] [Indexed: 12/17/2022] Open
Abstract
Ectopic lymphoid tissue, also known as tertiary lymphoid organs (TLO) develop adaptively within sites of chronic tissue inflammation, thereby allowing the host to efficiently crossprime specific immune effector cells within sites of disease. Recent evidence suggests that the presence of TLO in the tumor microenvironment (TME) predicts better overall survival. We will discuss the relevance of extranodal T cell priming within the TME as a means to effectively promote anti-tumor immunity and the strategic use of dendritic cell (DC)-based therapies to reinforce this clinically preferred process in the cancer-bearing host.
Collapse
Affiliation(s)
- Lu Chen
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Jennifer L Taylor
- Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA ; Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA ; University of Pittsburgh Cancer Institute , Pittsburgh, PA , USA
| |
Collapse
|
34
|
Abstract
A growing body of evidence suggests that non-hematopoietic stromal cells of the intestine have multiple roles in immune responses and inflammation at this mucosal site. Despite this, many still consider gut stromal cells as passive structural entities, with past research focused heavily on their roles in fibrosis, tumor progression, and wound healing, rather than their contributions to immune function. In this review, we discuss our current knowledge of stromal cells in intestinal immunity, highlighting the many immunological axes in which stromal cells have a functional role. We also consider emerging data that broaden the potential scope of their contribution to immunity in the gut and argue that these so-called "non-immune" cells are reclassified in light of their diverse contributions to intestinal innate immunity and the maintenance of mucosal homeostasis.
Collapse
|
35
|
Progression pattern of restrictive allograft syndrome after lung transplantation. J Heart Lung Transplant 2013; 32:23-30. [DOI: 10.1016/j.healun.2012.09.026] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Revised: 08/12/2012] [Accepted: 09/15/2012] [Indexed: 11/20/2022] Open
|
36
|
Sato M. Chronic lung allograft dysfunction after lung transplantation: the moving target. Gen Thorac Cardiovasc Surg 2012; 61:67-78. [DOI: 10.1007/s11748-012-0167-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Indexed: 11/29/2022]
|
37
|
Rejection of Tracheal Allograft by Intrapulmonary Lymphoid Neogenesis in the Absence of Secondary Lymphoid Organs. Transplantation 2012; 93:1212-20. [DOI: 10.1097/tp.0b013e318250fbf5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
38
|
Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol 2012; 33:297-305. [PMID: 22622061 PMCID: PMC7106385 DOI: 10.1016/j.it.2012.04.006] [Citation(s) in RCA: 299] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 04/17/2012] [Accepted: 04/17/2012] [Indexed: 01/07/2023]
Abstract
The lymph nodes (LNs) and spleen have an optimal structure that allows the interaction between T cells, B cells and antigen-presenting dendritic cells (DCs) on a matrix made up by stromal cells. Such a highly organized structure can also be formed in tertiary lymphoid organs (TLOs) at sites of infection or chronic immune stimulation. This review focuses on the molecular mechanisms of TLO formation and maintenance, the controversies surrounding the nature of the inducing events, and the functions of these structures in infection, transplantation and autoimmunity.
Collapse
|