1
|
Priya K, Rawat S, Das D, Chaubey M, Thacker H, Giri K, Singh S, Rai M, Mohanty S, Rai G. Autoimmunity and clinical pathology amelioration in SLE by dexamethasone primed mesenchymal stem cell derived conditioned media. Stem Cell Res Ther 2025; 16:158. [PMID: 40158179 PMCID: PMC11954324 DOI: 10.1186/s13287-025-04208-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/29/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND This study aimed to investigate the therapeutic potential of cell-free Dexamethasone (Dex) primed Wharton's jelly Mesenchymal stem cells derived conditioned media (DW) in addressing complications associated with systemic lupus erythematosus (SLE), focusing on its immunomodulatory effects. METHODS Peripheral blood mononuclear cells from 74 SLE patients were stimulated and treated with Dex, DW and W. Culture supernatant were evaluated for autoantibody levels, IL-10 and TGF-β by ELISA, Treg subtypes, Breg subtypes, TH17 cells Double negative T cells and inflammatory neutrophils by flow cytometry, IL-10 and IL-17A by qPCR. In vivo studies were performed on 60 pristane induced female BALB/c mice. Dex and DW treatments were evaluated for autoantibody production, proteinuria, immunomodulation of immune cells, organ function, and histopathology. In vivo imaging of internal organs was done using VevoLAZR-X photoacoustic imaging system. RESULTS DW treatment significantly expanded different Treg and Bregs subtypes. DW suppressed pathogenic TH17, Double negative T cells and inflammatory neutrophils. Comparative analyses with hydroxychloroquine showed similar effects, with combined treatment enhancing efficacy. Inhibition studies implicated the TGF-β pathway in DW's mechanism. In vivo studies using the PIL mouse model showed that DW treatment reduced mortality, prevented proteinuria, and ameliorated symptoms such as limb inflammation, seizures, and alopecia. Detailed organ-specific evaluations through live imaging and histopathological analyses revealed DW's protective effects on kidneys, liver, lungs, heart, and spleen. CONCLUSION DW shows promise as a cell-free biological therapy for SLE and related autoimmune disorders, capable of modulating immune responses effectively without the adverse effects of glucocorticoids.
Collapse
Affiliation(s)
- Khushbu Priya
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Sonali Rawat
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, AIIMS, New Delhi, 110029, India
| | - Doli Das
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Manaswi Chaubey
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Hiral Thacker
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Kiran Giri
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | | | - Madhukar Rai
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, AIIMS, New Delhi, 110029, India
| | - Geeta Rai
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
2
|
Li Y, Duan Q, Wang C, Du L, Jiang Z, Li S, Ruan X, Huang L, He Z, Wen C, Zhang Y. Jieduquyuziyin prescription alleviates lupus development via inhibiting neddylation pathway to promote Bim-induced apoptosis of double negative T cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118884. [PMID: 39362327 DOI: 10.1016/j.jep.2024.118884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jieduquyuziyin prescription (JP) is an empirical prescription approved for application to treat systemic lupus erythematosus (SLE) in hospital within China. Despite the prominent treatment effect of JP clinically, further investigation is imperative to explore its underlying mechanisms. AIM OF THE STUDY We aim to investigate the impact of JP on DN T cell apoptosis in the treatment of SLE and the specific regulation mechanisms. MATERIALS AND METHODS Firstly, female MRL/lpr mice were treated with JP and the therapeutic efficacy of JP was evaluated via skin lesions, lymphoid organ enlargement, accumulation of autoantibodies and renal function. Then, flow cytometer analysis was performed to evaluate the proportions and the apoptosis of T cell subpopulations. Based on the above results, double-negative (DN) T cells were subjected to proteomic with subsequent differential screening. The expression of Ube2m and Bim was further validated using real-time PCR and Western blot. Subsequently, DN T cells were incubated with JP-contained serum in vitro, and cell apoptosis was quantified using flow cytometry. Additionally, the expression levels of Ube2m, Bim and other associated proteins were also assessed through western blotting. To further clarify whether Ube2m serves as the key target of JP in regulating DN T cell apoptosis, the mice that Ube2m was specific deleted in T cells with spontaneous lupus (Ube2m-/-lpr) were utilized. JP was administered to WTlpr or Ube2m-/-lpr mice, followed by assessment of the lupus condition and DN T cell apoptosis. RESULTS JP administration effectively ameliorated the lupus phenotype. Then flow cytometry assay showed that JP treatment enhanced DN T cell apoptosis to reduce their accumulation and restored the immune homeostasis. Proteomic analysis revealed a significant inhibition of Ube2m for JP treatment, which is essential for maintaining homeostasis of DN T cells. Further experiments confirmed that JP treatment effectively downregulated the expression of Ube2m and subsequently upregulated the level of pro-apoptotic protein Bim with decreased Bim degradation. In vitro experiments also confirmed that JP-contained serum significantly facilitated DN T cell apoptosis and reduced DN T cell accumulation by inhibiting Ube2m expression. Furthermore, Ube2m-/-lpr mice were utilized and the impact of JP treatment on the apoptosis of DN T cells was found to be minimal in the absence of Ube2m. Mechanistic investigation reveals that JP exerts its effects by suppressing the expression of Ube2m, subsequently inhibiting CRL-dependent degradation of Bim, and ultimately promoting Bim-induced apoptosis in DN T cells. Furthermore, the blockade of Ube2m in T cells effectively prevents JP-induced apoptosis in DN T cells, underscoring Ube2m as one crucial therapeutic target of JP in mediating DN T cell apoptosis and managing SLE. CONCLUSIONS Our findings indicate that JP treatment effectively restores the homeostasis of DN T cells in SLE by inhibiting Ube2m expression, thereby reducing Bim ubiquitination degradation. This ultimately enhanced DN T cell apoptosis and alleviated lupus phenotype.
Collapse
Affiliation(s)
- Yiping Li
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Qingchi Duan
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Chenxi Wang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Lijun Du
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Zhangsheng Jiang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Suling Li
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xinyi Ruan
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Lin Huang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zhixing He
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Chengping Wen
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yun Zhang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
3
|
Dossybayeva K, Zhubanova G, Mussayeva A, Mukusheva Z, Dildabayeva A, Nauryzbayeva G, Akhmaltdinova L, Orumbayeva U, Tanko M, Poddighe D. Nonspecific increase of αβTCR + double-negative T cells in pediatric rheumatic diseases. World J Pediatr 2024; 20:1283-1292. [PMID: 39604769 PMCID: PMC11634929 DOI: 10.1007/s12519-024-00854-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND An increased number of double-negative T (DNT) cells expressing the αβ T cell receptor (αβ+DNT cells) is one of the diagnostic criteria for autoimmune lymphoproliferative syndrome (ALPS). Moreover, these cells are expanded in a widely used murine model for lupus. However, the homeostasis of αβ+DNT cells remains inadequately investigated in rheumatic disorders, especially in pediatric patients. METHODS In this cross-sectional, prospective, and observational study, children with rheumatic disorders and healthy controls were recruited to analyze the quantity and characteristics of circulating DNT cells using flow cytometry. RESULTS Overall, the two study groups did not differ in their total DNT cell pool in the bloodstream. However, the number of αβ+DNT cells was significantly higher in rheumatic children than that in the controls, whereas the γδ+DNT cells remained similar. This expansion in the circulating pool of αβ+DNT cells was comparable across different rheumatic diseases, all showing significant differences from the controls in this regard. Moreover, no significant correlation was found between αβ+DNT cell numbers and disease activity. CONCLUSIONS These preliminary results indicate that circulating αβ+DNT cells are significantly expanded in children with rheumatic disorders; however, this finding appears to be a nonspecific (disease-unrelated) marker of autoimmunity. Further and larger studies are necessary to better investigate and define the role of DNT cells in pediatric rheumatic diseases.
Collapse
Affiliation(s)
- Kuanysh Dossybayeva
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan
| | - Gulsamal Zhubanova
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan
| | - Assel Mussayeva
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan
| | - Zaure Mukusheva
- Program of Pediatric Rheumatology, Clinical Academic Department of Pediatrics, University Medical Center, Astana, Kazakhstan
| | - Aiken Dildabayeva
- Program of Pediatric Rheumatology, Clinical Academic Department of Pediatrics, University Medical Center, Astana, Kazakhstan
| | - Galiya Nauryzbayeva
- Clinical Academic Department of Laboratory Medicine, Pathology and Genetics, Republican Diagnostic Center, University Medical Center, Astana, Kazakhstan
| | - Lyudmila Akhmaltdinova
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health (NRCMCH), University Medical Center(UMC), 010000, Astana, Kazakhstan
| | - Ulbolsyn Orumbayeva
- Clinical Academic Department of Laboratory Medicine, Pathology and Genetics, Republican Diagnostic Center, University Medical Center, Astana, Kazakhstan
| | - Matthew Tanko
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan
- Clinical Academic Department of Laboratory Medicine, Pathology and Genetics, Republican Diagnostic Center, University Medical Center, Astana, Kazakhstan
| | - Dimitri Poddighe
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan.
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health (NRCMCH), University Medical Center(UMC), 010000, Astana, Kazakhstan.
- College of Health Sciences, VinUniversity, Gia Lam District, Hanoi, Vietnam.
| |
Collapse
|
4
|
Poddighe D, Maulenkul T, Dossybayeva K, Zhubanova G, Mukusheva Z, Akhmaltdinova L. Double-negative T cells in pediatric rheumatic diseases. Clin Exp Pediatr 2024; 67:632-640. [PMID: 39265625 PMCID: PMC11621738 DOI: 10.3345/cep.2023.01760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 09/14/2024] Open
Abstract
Double-negative (CD4-CD8-) T (DNT) cells have been implicated in autoimmune lymphoproliferative syndrome (ALPS), where their expansion inside the circulating pool of T cells represents a diagnostic criterion. Recent experimental evidence has supported the immunomodulatory roles of DNT cells, and studies in adult patients have suggested that they may be altered in some immune-mediated conditions. This study aimed to retrieve available data on circulating DNT cells in pediatric rheumatic disorders that do not arise in the context of ALPS through a systematic literature review of 3 scientific databases (PubMed, Scopus, and Web of Science). The final output of the systematic literature search consisted of 8 manuscripts, including cross-sectional (n=6) and longitudinal (n=2) studies. Overall, the pooled population of patients includes children affected with pediatric systemic lupus erythematosus (n= 104), juvenile idiopathic arthritis (n=92), Behçet's disease (n=15), mixed connective tissue disease (n=8), juvenile dermatomyositis (n=6), and Kawasaki disease/multisystem inflammatory disease in children (n=1 and n=14, respectively); moreover, one study also included 11 children with a high titer of antinuclear antibody but no diagnosis of rheumatic disease. All studies except one included a control group. The number of DNT cells were increased in most studies of children with rheumatic diseases. Even if such a limited number of studies and their great heterogeneity in several methodological aspects do not allow for reliable conclusions about the relevance of DNT cells in specific rheumatic conditions in children, this cell population deserves further investigation in this pathological setting through well-designed clinical studies.
Collapse
Affiliation(s)
- Dimitri Poddighe
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana, Kazakhstan
- College of Health Sciences, VinUniversity, Hanoi, Vietnam
| | - Tilektes Maulenkul
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana, Kazakhstan
| | - Kuanysh Dossybayeva
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
| | - Gulsamal Zhubanova
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
| | - Zaure Mukusheva
- Program of Pediatric Rheumatology, Clinical Academic Department of Pediatrics, University Medical Center, Astana, Kazakhstan
| | - Lyudmila Akhmaltdinova
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
| |
Collapse
|
5
|
Zhang Y, Du L, Wang C, Jiang Z, Duan Q, Li Y, Xie Z, He Z, Sun Y, Huang L, Lu L, Wen C. Neddylation is a novel therapeutic target for lupus by regulating double negative T cell homeostasis. Signal Transduct Target Ther 2024; 9:18. [PMID: 38221551 PMCID: PMC10788348 DOI: 10.1038/s41392-023-01709-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 10/15/2023] [Accepted: 11/15/2023] [Indexed: 01/16/2024] Open
Abstract
Systemic lupus erythematosus (SLE), a severe autoimmune disorder, is characterized by systemic inflammatory response, autoantibody accumulation and damage to organs. The dysregulation of double-negative (DN) T cells is considered as a crucial commander during SLE. Neddylation, a significant type of protein post-translational modification (PTM), has been well-proved to regulate T cell-mediated immune response. However, the function of neddylation in SLE is still unknown. Here, we reported that neddylation inactivation with MLN4924, a specific inhibitor of NEDD8-activating enzyme E1 (NAE1), or genetic abrogation of Ube2m in T cells decreased DN T cell accumulation and attenuated murine lupus development. Further investigations revealed that inactivation of neddylation blocked Bim ubiquitination degradation and maintained Bim level in DN T cells, contributing to the apoptosis of the accumulated DN T cells in lupus mice. Then double knockout (KO) lupus-prone mice (Ube2m-/-Bim-/-lpr) were generated and results showed that loss of Bim reduced Ube2m deficiency-induced apoptosis in DN T cells and reversed the alleviated lupus progression. Our findings identified that neddylation inactivation promoted Bim-mediated DN T cell apoptosis and attenuated lupus progression. Clinically, we also found that in SLE patients, the proportion of DN T cells was raised and their apoptosis was reduced. Moreover, compared to healthy groups, SLE patients exhibited decreased Bim levels and elevated Cullin1 neddylation levels. Meantime, the inhibition of neddylation induced Bim-dependent apoptosis of DN T cells isolated from SLE patients. Altogether, our findings provide the direct evidence about the function of neddylation during lupus, suggesting a promising therapeutic approach for this disease.
Collapse
Affiliation(s)
- Yun Zhang
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lijun Du
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Chenxi Wang
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zhangsheng Jiang
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qingchi Duan
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yiping Li
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zhijun Xie
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zhixing He
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yi Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
- Cancer Center of Zhejiang University, Hangzhou, 310029, China
| | - Lin Huang
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China.
- Chongqing International Institute for Immunology, Chongqing, 400038, China.
| | - Chengping Wen
- Key Laboratory of Chinese medicine rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
6
|
Poddighe D, Dossybayeva K, Kozhakhmetov S, Rozenson R, Assylbekova M. Double-Negative T (DNT) Cells in Patients with Systemic Lupus Erythematosus. Biomedicines 2024; 12:166. [PMID: 38255272 PMCID: PMC10812956 DOI: 10.3390/biomedicines12010166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Double-negative T (DNT) cells are a rare and unconventional T-lymphocyte subpopulation lacking both CD4 and CD8 markers. Their immunopathological roles and clinical relevance have yet to be elucidated. Beyond autoimmune lymphoproliferative syndrome (ALPS), these cells may also play a role in rheumatic disorders, including systemic lupus erythematosus (SLE); indeed, these two diseases share several autoimmune manifestations (including nephritis). Moreover, one of the main experimental murine models used to investigate lupus, namely the MRL/lpr mouse, is characterized by an expansion of DNT cells, which can support the production of pathogenic autoantibodies and/or modulate the immune response in this context. However, lupus murine models are not completely consistent with their human SLE counterpart, of course. In this mini review, we summarize and analyze the most relevant clinical studies investigating the DNT cell population in SLE patients. Overall, based on the present literature review and analysis, DNT cell homeostasis seems to be altered in patients with SLE. Indeed, most of the available clinical studies (which include both adults and children) reported an increased DNT cell percentage in SLE patients, especially during the active phases, even though no clear correlation with disease activity and/or inflammatory parameters has been clearly established. Well-designed, standardized, and longitudinal clinical studies focused on DNT cell population are needed, in order to further elucidate the actual contribution of these cells in SLE pathogenesis and their interactions with other immune cells (also implicated and/or altered in SLE, such as basophils), and clarify whether their expansion and/or immunophenotypic aspects may have any immunopathological relevance (and, then, represent potential disease markers and, in perspective, even therapeutic targets) or are just an unspecific epiphenomenon of autoimmunity.
Collapse
Affiliation(s)
- Dimitri Poddighe
- School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan;
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana 010000, Kazakhstan;
| | | | - Samat Kozhakhmetov
- Center for Life Science, National Laboratory Astana, Astana 010000, Kazakhstan;
| | - Rafail Rozenson
- Department of Children’s Diseases n.1, Astana Medical University, Astana 010000, Kazakhstan;
| | - Maykesh Assylbekova
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana 010000, Kazakhstan;
| |
Collapse
|
7
|
Verweyen EL, Thakkar K, Dhakal S, Baker E, Chetal K, Schnell D, Canna S, Grom AA, Salomonis N, Schulert GS. Population-level single-cell genomics reveals conserved gene programs in systemic juvenile idiopathic arthritis. J Clin Invest 2023; 133:e166741. [PMID: 37733441 PMCID: PMC10645394 DOI: 10.1172/jci166741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 09/19/2023] [Indexed: 09/23/2023] Open
Abstract
Systemic autoimmune and autoinflammatory diseases are characterized by genetic and cellular heterogeneity. While current single-cell genomics methods provide insights into known disease subtypes, these analysis methods do not readily reveal novel cell-type perturbation programs shared among distinct patient subsets. Here, we performed single-cell RNA-Seq of PBMCs of patients with systemic juvenile idiopathic arthritis (SJIA) with diverse clinical manifestations, including macrophage activation syndrome (MAS) and lung disease (LD). We introduced two new computational frameworks called UDON and SATAY-UDON, which define patient subtypes based on their underlying disrupted cellular programs as well as associated biomarkers or clinical features. Among twelve independently identified subtypes, this analysis uncovered what we believe to be a novel complement and interferon activation program identified in SJIA-LD monocytes. Extending these analyses to adult and pediatric lupus patients found new but also shared disease programs with SJIA, including interferon and complement activation. Finally, supervised comparison of these programs in a compiled single-cell pan-immune atlas of over 1,000 healthy donors found a handful of normal healthy donors with evidence of early inflammatory activation in subsets of monocytes and platelets, nominating possible biomarkers for early disease detection. Thus, integrative pan-immune single-cell analysis resolved what we believe to be new conserved gene programs underlying inflammatory disease pathogenesis and associated complications.
Collapse
Affiliation(s)
| | - Kairavee Thakkar
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Daniel Schnell
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Scott Canna
- Children’s Hospital of Philadelphia, Division of Rheumatology, Philadelphia, Pennsylvania, USA
| | - Alexei A. Grom
- Division of Rheumatology and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Grant S. Schulert
- Division of Rheumatology and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
8
|
Failing C, Blase JR, Walkovich K. Understanding the Spectrum of Immune Dysregulation Manifestations in Autoimmune Lymphoproliferative Syndrome and Autoimmune Lymphoproliferative Syndrome-like Disorders. Rheum Dis Clin North Am 2023; 49:841-860. [PMID: 37821199 DOI: 10.1016/j.rdc.2023.07.001] [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: 10/13/2023]
Abstract
As a disorder of immune dysregulation, autoimmune lymphoproliferative syndrome (ALPS) stems from pathogenic variants in the first apoptosis signal-mediated apoptosis (Fas) and Fas-ligand pathway that result in elevations of CD3+ TCRαβ+ CD4- CD8- T cells along with chronic lymphoproliferation, a heightened risk for malignancy, and importantly for the rheumatologist, increased risk of autoimmunity. While immune cytopenias are the most encountered autoimmune phenomena, there is increasing appreciation for ocular, musculoskeletal, pulmonary and renal inflammatory manifestations similar to more common rheumatology diseases. Additionally, ALPS-like conditions that share similar clinical features and opportunities for targeted therapy are increasingly recognized via genetic testing, highlighting the need for rheumatologists to be facile in the recognition and diagnosis of this spectrum of disorders. This review will focus on clinical and laboratory features of both ALPS and ALPS-like disorders with the intent to provide a framework for rheumatologists to understand the pathophysiologic drivers and discriminate between diagnoses.
Collapse
Affiliation(s)
- Christopher Failing
- Sanford Health, Fargo, ND, USA; University of North Dakota School of Medicine and Health Sciences, Grand Folks, ND, USA.
| | - Jennifer R Blase
- University of Michigan, 1500 East Medical Center Drive, D4202 Medical Professional Building, Ann Arbor, MI 48109, USA
| | - Kelly Walkovich
- University of Michigan, 1500 East Medical Center Drive, D4202 Medical Professional Building, Ann Arbor, MI 48109, USA
| |
Collapse
|
9
|
Han C, Sheng Y, Wang J, Zhou X, Li W, Zhang C, Guo L, Yang Y. Double-negative T cells mediate M1 polarization of microglial cells via TNF-α-NLRP3 to aggravate neuroinflammation and cognitive impairment in Alzheimer's disease mice. J Cell Physiol 2022; 237:3860-3871. [PMID: 35866513 DOI: 10.1002/jcp.30839] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/03/2022] [Accepted: 07/12/2022] [Indexed: 11/09/2022]
Abstract
We mainly study the role and regulatory mechanism of double-negative T cells (DNTs) in Alzheimer's disease (AD). The mice splenic DNTs were separated and amplified by Rosettesep antibody adsorption method and Easysep magnetic activated cell sorting. DNTs were intraperitoneally injected into the APP/PS1-AD mice model, which was found to aggravate cognitive impairment in mice. DNTs secreted tumor necrosis factor α (TNF-α) to promote the activation of NLRP3 and the M1 polarization of microglial cells, and silencing NLRP3 with small interfering RNA (siRNA) suppressed the effect of DNTs. DNTs were later cocultured with mice microglial cell line BV2, then fluorescence staining was conducted to detect NLRP3 expression, and enzyme-linked immunoassay was performed to measure the expression of inflammatory factors. Moreover, the levels of NLRP3, ASC, and TNFR1 proteins were detected by western-blot assay, and the proportion of F4/80 + CD11b + M1 cells was detected by flow cytometry. DNTs promoted the M1 polarization of BV2 cells and the activation of NLRP3 inflammasome. After treatment of BV2 cells with NLRP3 inhibitor, the effect of DNTs was weakened. Later, TNF-α siRNA was transfected into DNTs, and it was found that DNTs with TNF-α silencing had markedly weakened polarization effect on BV2 cells. We discovered that the proportion of DNTs increased in AD patients. DNTs secreted TNF-α to regulate the activation of NLRP3 inflammasome and the M1 polarization of microglial cells, thus promoting the central inflammatory response and aggravating the cognitive impairment in AD mice.
Collapse
Affiliation(s)
- Chenyang Han
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yongjia Sheng
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jin Wang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaohong Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Wenyan Li
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Caiqun Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Li Guo
- Department of Center Laboratory, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yi Yang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
10
|
Velikkakam T, Gollob KJ, Dutra WO. Double-negative T cells: Setting the stage for disease control or progression. Immunology 2022; 165:371-385. [PMID: 34939192 PMCID: PMC10626195 DOI: 10.1111/imm.13441] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 12/15/2021] [Indexed: 11/30/2022] Open
Abstract
Double-negative (DN) T cells are present at relatively low frequencies in human peripheral blood, and are characterized as expressing the alpha-beta or gamma-delta T-cell receptor (TCR), but not the CD4 nor the CD8 co-receptors. Despite their low frequencies, these cells are potent producers of cytokines and, thus, are key orchestrators of immune responses. DN T cells were initially associated with induction of peripheral immunological tolerance and immunomodulatory activities related to disease prevention. However, other studies demonstrated that these cells can also display effector functions associated with pathology development. This apparent contradiction highlighted the heterogeneity of the DN T-cell population. Here, we review phenotypic and functional characteristics of DN T cells, emphasizing their role in human diseases. The need for developing biomarkers to facilitate the translation of studies from animal models to humans will also be discussed. Finally, we will examine DN T cells as promising therapeutic targets to prevent or inhibit human disease development.
Collapse
Affiliation(s)
- Teresiama Velikkakam
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Pós-graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kenneth J. Gollob
- Hospital Israelita Albert Einsten, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais – INCT-DT, Belo Horizonte, Brazil
| | - Walderez Ornelas Dutra
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Pós-graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais – INCT-DT, Belo Horizonte, Brazil
| |
Collapse
|
11
|
Flinn AM, Gennery AR. Primary immune regulatory disorders: Undiagnosed needles in the haystack? Orphanet J Rare Dis 2022; 17:99. [PMID: 35241125 PMCID: PMC8895571 DOI: 10.1186/s13023-022-02249-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 02/13/2022] [Indexed: 12/16/2022] Open
Abstract
Primary Immune Regulatory Disorders (PIRD) describe a group of conditions characterized by loss of normal inflammatory control and immune tolerance mechanisms, with autoimmunity as a predominant clinical feature. PIRD can arise due to defects in the number or function of regulatory T-lymphocytes, defects in the immune mechanisms required to ‘turn off’ inflammation such as in perforin-dependent cytotoxicity or alterations in cytokine signalling pathways. Diagnosis of PIRD is a significant challenge to physicians due to their rarity, complexity, and diversity in clinical manifestations. Many of these individual conditions lack a genotype–phenotype correlation and display incomplete penetrance. However, establishing a diagnosis is integral in optimizing patient management, including the use of individualized treatment approaches. Increasing awareness among physicians is necessary as patients are likely to present to different subspecialties. Due to the rarity of these conditions, worldwide collaboration and data-sharing is essential to improve our knowledge of the clinical spectrum and disease course in PIRD, and to optimize therapeutic strategies including identification of which patients can benefit from hematopoietic stem cell transplant.
Collapse
Affiliation(s)
- Aisling M Flinn
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
12
|
Schiavo E, Martini B, Attardi E, Consonni F, Ciullini Mannurita S, Coniglio ML, Tellini M, Chiocca E, Fotzi I, Luti L, D'Alba I, Veltroni M, Favre C, Gambineri E. Autoimmune Cytopenias and Dysregulated Immunophenotype Act as Warning Signs of Inborn Errors of Immunity: Results From a Prospective Study. Front Immunol 2022; 12:790455. [PMID: 35058929 PMCID: PMC8765341 DOI: 10.3389/fimmu.2021.790455] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/22/2021] [Indexed: 12/19/2022] Open
Abstract
Inborn errors of immunity (IEI) are genetic disorders characterized by a wide spectrum of clinical manifestations, ranging from increased susceptibility to infections to significant immune dysregulation. Among these, primary immune regulatory disorders (PIRDs) are mainly presenting with autoimmune manifestations, and autoimmune cytopenias (AICs) can be the first clinical sign. Significantly, AICs in patients with IEI often fail to respond to first-line therapy. In pediatric patients, autoimmune cytopenias can be red flags for IEI. However, for these cases precise indicators or parameters useful to suspect and screen for a hidden congenital immune defect are lacking. Therefore, we focused on chronic/refractory AIC patients to perform an extensive clinical evaluation and multiparametric flow cytometry analysis to select patients in whom PIRD was strongly suspected as candidates for genetic analysis. Key IEI-associated alterations causative of STAT3 GOF disease, IKAROS haploinsufficiency, activated PI3Kδ syndrome (APDS), Kabuki syndrome and autoimmune lymphoproliferative syndrome (ALPS) were identified. In this scenario, a dysregulated immunophenotype acted as a potential screening tool for an early IEI diagnosis, pivotal for appropriate clinical management and for the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Ebe Schiavo
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Beatrice Martini
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Enrico Attardi
- Division of Hematology, Careggi University Hospital, Florence, Italy
| | - Filippo Consonni
- Meyer University Children's Hospital, University of Florence, Florence, Italy
| | - Sara Ciullini Mannurita
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Maria Luisa Coniglio
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Marco Tellini
- Meyer University Children's Hospital, University of Florence, Florence, Italy
| | - Elena Chiocca
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Ilaria Fotzi
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Laura Luti
- Division of Pediatric Oncology/Hematology, University Hospital of Pisa, Pisa, Italy
| | - Irene D'Alba
- Division of Pediatric Oncology/Hematology, University Hospital of Ospedali Riuniti, Ancona, Italy
| | - Marinella Veltroni
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Claudio Favre
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | - Eleonora Gambineri
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy.,Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| |
Collapse
|
13
|
Consonni F, Gambineri E, Favre C. ALPS, FAS, and beyond: from inborn errors of immunity to acquired immunodeficiencies. Ann Hematol 2022; 101:469-484. [PMID: 35059842 PMCID: PMC8810460 DOI: 10.1007/s00277-022-04761-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022]
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a primary immune regulatory disorder characterized by benign or malignant lymphoproliferation and autoimmunity. Classically, ALPS is due to mutations in FAS and other related genes; however, recent research revealed that other genes could be responsible for similar clinical features. Therefore, ALPS classification and diagnostic criteria have changed over time, and several ALPS-like disorders have been recently identified. Moreover, mutations in FAS often show an incomplete penetrance, and certain genotypes have been associated to a dominant or recessive inheritance pattern. FAS mutations may also be acquired or could become pathogenic when associated to variants in other genes, delineating a possible digenic type of inheritance. Intriguingly, variants in FAS and increased TCR αβ double-negative T cells (DNTs, a hallmark of ALPS) have been identified in multifactorial autoimmune diseases, while FAS itself could play a potential role in carcinogenesis. These findings suggest that alterations of FAS-mediated apoptosis could trespass the universe of inborn errors of immunity and that somatic mutations leading to ALPS could only be the tip of the iceberg of acquired immunodeficiencies.
Collapse
Affiliation(s)
- Filippo Consonni
- Anna Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Eleonora Gambineri
- Division of Pediatric Oncology/Hematology, BMT Unit, Meyer University Children's Hospital, Viale Gaetano Pieraccini 24, 50139, Florence, Italy.
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy.
| | - Claudio Favre
- Division of Pediatric Oncology/Hematology, BMT Unit, Meyer University Children's Hospital, Viale Gaetano Pieraccini 24, 50139, Florence, Italy
| |
Collapse
|
14
|
Rais A, Mekki N, Fedhila F, Alosaimi MF, Ben Khaled M, Zameli A, Agrebi N, Sellami MK, Geha R, Ben-Mustapha I, Barbouche MR. Case Report: FOXP3 Mutation in a Patient Presenting With ALPS. Front Immunol 2021; 12:692107. [PMID: 34531853 PMCID: PMC8438314 DOI: 10.3389/fimmu.2021.692107] [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] [Received: 04/07/2021] [Accepted: 07/19/2021] [Indexed: 01/21/2023] Open
Abstract
ALPS and IPEX are two well-characterized inborn errors of immunity with immune dysregulation, considered as two master models of monogenic auto-immune diseases. Thus, with autoimmunity as their primary clinical manifestation, these two entities may show clinical overlap. Traditionally, immunological biomarkers are used to establish an accurate differential diagnosis. Herein, we describe a patient who presented with clinical features and biomarkers fulfilling the diagnostic criteria of ALPS. Severe apoptotic defect was also shown in the patient's cell lines and PHA-activated peripheral blood lymphocytes. Sanger sequencing of the FAS gene did not reveal any causal mutation. NGS screening revealed a novel deleterious variant located in the N terminal repressor domain of FOXP3 but no mutations in the FAS pathway-related genes. TEMRA cells (terminally differentiated effector memory cells re-expressing CD45RA) and PD1 expression were increased arguing in favor of T-cell exhaustion, which could be induced by unrestrained activation of T effector cells because of Treg deficiency. Moreover, defective FOXP3 observed in the patient could intrinsically induce increased proliferation and resistance to apoptosis in T effector cells. This observation expands the spectrum of FOXP3 deficiency and underscores the role of NGS in detecting mutations that induce overlapping phenotypes among inborn errors of immunity with immune dysregulation. In addition, these findings suggest a potential link between FOXP3 and FAS pathways.
Collapse
Affiliation(s)
- Afef Rais
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Medicine, Université de Tunis El Manar, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia
| | - Najla Mekki
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Medicine, Université de Tunis El Manar, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia
| | - Faten Fedhila
- Faculty of Medicine, Université de Tunis El Manar, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia.,Department of Pediatrics A, Children's Hospital, Tunis, Tunisia
| | | | - Monia Ben Khaled
- Faculty of Medicine, Université de Tunis El Manar, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia.,Pediatric Immuno-Hematology unit, Bone Marrow Transplantation Center Tunis, Tunis, Tunisia
| | - Amal Zameli
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia
| | - Nourhen Agrebi
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia
| | - Maryam Kallel Sellami
- Faculty of Medicine, Université de Tunis El Manar, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia.,Department of Immunology, La Rabta University Hospital, Tunis, Tunisia
| | - Raif Geha
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Imen Ben-Mustapha
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Medicine, Université de Tunis El Manar, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia
| | - Mohamed-Ridha Barbouche
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Medicine, Université de Tunis El Manar, Tunis, Tunisia.,Université de Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
15
|
Aluri J, Bach A, Kaviany S, Chiquetto Paracatu L, Kitcharoensakkul M, Walkiewicz MA, Putnam CD, Shinawi M, Saucier N, Rizzi EM, Harmon MT, Keppel MP, Ritter M, Similuk M, Kulm E, Joyce M, de Jesus AA, Goldbach-Mansky R, Lee YS, Cella M, Kendall PL, Dinauer MC, Bednarski JJ, Bemrich-Stolz C, Canna SW, Abraham SM, Demczko MM, Powell J, Jones SM, Scurlock AM, De Ravin SS, Bleesing JJ, Connelly JA, Rao VK, Schuettpelz LG, Cooper MA. Immunodeficiency and bone marrow failure with mosaic and germline TLR8 gain of function. Blood 2021; 137:2450-2462. [PMID: 33512449 PMCID: PMC8109013 DOI: 10.1182/blood.2020009620] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022] Open
Abstract
Inborn errors of immunity (IEI) are a genetically heterogeneous group of disorders with a broad clinical spectrum. Identification of molecular and functional bases of these disorders is important for diagnosis, treatment, and an understanding of the human immune response. We identified 6 unrelated males with neutropenia, infections, lymphoproliferation, humoral immune defects, and in some cases bone marrow failure associated with 3 different variants in the X-linked gene TLR8, encoding the endosomal Toll-like receptor 8 (TLR8). Interestingly, 5 patients had somatic variants in TLR8 with <30% mosaicism, suggesting a dominant mechanism responsible for the clinical phenotype. Mosaicism was also detected in skin-derived fibroblasts in 3 patients, demonstrating that mutations were not limited to the hematopoietic compartment. All patients had refractory chronic neutropenia, and 3 patients underwent allogeneic hematopoietic cell transplantation. All variants conferred gain of function to TLR8 protein, and immune phenotyping demonstrated a proinflammatory phenotype with activated T cells and elevated serum cytokines associated with impaired B-cell maturation. Differentiation of myeloid cells from patient-derived induced pluripotent stem cells demonstrated increased responsiveness to TLR8. Together, these findings demonstrate that gain-of-function variants in TLR8 lead to a novel childhood-onset IEI with lymphoproliferation, neutropenia, infectious susceptibility, B- and T-cell defects, and in some cases, bone marrow failure. Somatic mosaicism is a prominent molecular mechanism of this new disease.
Collapse
Affiliation(s)
| | - Alicia Bach
- Division of Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Saara Kaviany
- Pediatric Hematology Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Luana Chiquetto Paracatu
- Division of Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Maleewan Kitcharoensakkul
- Division of Rheumatology/Immunology and
- Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Magdalena A Walkiewicz
- Centralized Sequencing Initiative, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Christopher D Putnam
- Department of Medicine, University of California School of Medicine, San Diego, La Jolla, CA
- San Diego Branch, Ludwig Institute for Cancer Research, La Jolla, CA
| | - Marwan Shinawi
- Division of Genetics and Genomic Medicine, Department of Pediatrics and
| | | | - Elise M Rizzi
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | | | | | - Morgan Similuk
- Centralized Sequencing Initiative, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Elaine Kulm
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD
| | | | - Adriana A de Jesus
- Translational Autoinflammatory Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Yi-Shan Lee
- Division of Anatomic and Molecular Pathology and
| | - Marina Cella
- Division of Immunology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Peggy L Kendall
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Division of Immunology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Mary C Dinauer
- Division of Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Jeffrey J Bednarski
- Division of Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Christina Bemrich-Stolz
- Division of Hematology and Oncology, Department of Pediatrics, University of Alabama School of Medicine, Birmingham, AL
| | - Scott W Canna
- Division of Pediatric Rheumatology and RK Mellon Institute, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh and University of Pittsburgh, Pittsburgh, PA
| | - Shirley M Abraham
- Division of Hematology and Oncology, Department of Pediatrics, University of New Mexico, Albuquerque, NM
| | | | - Jonathan Powell
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Nemours Alfred I. DuPont Hospital for Children, Wilmington, DE
| | - Stacie M Jones
- Division of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, AR
| | - Amy M Scurlock
- Division of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, AR
| | - Suk See De Ravin
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD; and
| | - Jack J Bleesing
- Division of Bone Marrow Transplantation and Immunodeficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - James A Connelly
- Pediatric Hematology Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - V Koneti Rao
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD; and
| | - Laura G Schuettpelz
- Division of Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | | |
Collapse
|
16
|
Oliveira Mendonça L, Matucci-Cerinic C, Terranova P, Casabona F, Bovis F, Caorsi R, Fioredda F, Palmisani E, Grossi A, Guardo D, Bustaffa M, Volpi S, Ceccherini I, Ravelli A, Dufour C, Miano M, Gattorno M. The challenge of early diagnosis of autoimmune lymphoproliferative syndrome in children with suspected autoinflammatory/autoimmune disorders. Rheumatology (Oxford) 2021; 61:696-704. [PMID: 33909886 DOI: 10.1093/rheumatology/keab361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/12/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To test the usefulness of an extended panel of lymphocyte subsets (LS) in combination with Oliveira's diagnostic criteria for the identification of autoimmune lymphoproliferative syndrome (ALPS) in children referred to a pediatric rheumatology center. METHODS patients referred from 2015 to 2018 to our Rheumatology Unit for an autoimmune or autoinflammatory condition were retrospectively analyzed. Oliveira's required criteria (chronic lymphoproliferation and elevated DNT) were applied as first screening. Flow cytometry study included double negative CD4-CD8-TCR αβ+T lymphocytes (DNT), CD25+CD3+, HLA-DR+CD3+T cells, B220+T cells, and CD27+B cells. Data were analyzed with an univariate logistic regression analysis, followed by a multivariate analysis. Sensitivity and specificity of the Oliveira's required criteria were calculated. RESULTS 264 patients were included in the study and classified as: i) autoimmune diseases (26); ii) juvenile idiopathic arthritis (JIA) (35) iii) monogenic systemic autoinflammatory disease (SAID) (27); iv) PFAPA syndrome (100); v) systemic undefined recurrent fever (SURF) (45); vi) undetermined-SAID (14); vii) ALPS (17). Oliveira's required criteria displayed a sensitivity of 100% and specificity of 79%. When compared with other diseases the TCRαβ+B220+ lymphocytes were significantly increased in ALPS patients. The multivariate analysis revealed 5 clinical/laboratory parameters positively associated to ALPS: splenomegaly, female gender, arthralgia, elevated DNT and TCRαβ+B220+lymphocytes. CONCLUSIONS Oliveira's required criteria are useful for the early suspicion of ALPS. TCRαβ+B220+ lymphocytes should be added in the diagnostic work-up of patients referred to pediatric rheumatology unit for a suspected autoimmune or autoinflammatory condition, providing a relevant support in the early diagnosis of ALPS.
Collapse
Affiliation(s)
| | - Caterina Matucci-Cerinic
- Clinic of Pediatrics and Rheumatology, IRCCS G. Gaslini and University of Genoa
- DINOGMI, University of Genoa
| | | | | | | | - Roberta Caorsi
- Center for Autoinflammatory Diseases and Immunodeficiencies, IRCCS G. Gaslini
| | | | | | - Alice Grossi
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS G. Gaslini, Genoa, Italy
| | | | - Marta Bustaffa
- Clinic of Pediatrics and Rheumatology, IRCCS G. Gaslini and University of Genoa
- DINOGMI, University of Genoa
| | - Stefano Volpi
- Center for Autoinflammatory Diseases and Immunodeficiencies, IRCCS G. Gaslini
- DINOGMI, University of Genoa
| | - Isabella Ceccherini
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS G. Gaslini, Genoa, Italy
| | - Angelo Ravelli
- Clinic of Pediatrics and Rheumatology, IRCCS G. Gaslini and University of Genoa
| | | | | | - Marco Gattorno
- Center for Autoinflammatory Diseases and Immunodeficiencies, IRCCS G. Gaslini
| |
Collapse
|
17
|
Papillion A, Ballesteros-Tato A. The Potential of Harnessing IL-2-Mediated Immunosuppression to Prevent Pathogenic B Cell Responses. Front Immunol 2021; 12:667342. [PMID: 33986755 PMCID: PMC8112607 DOI: 10.3389/fimmu.2021.667342] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/06/2021] [Indexed: 11/18/2022] Open
Abstract
Immunosuppressive drugs can partially control Antibody (Ab)-dependent pathology. However, these therapeutic regimens must be maintained for the patient's lifetime, which is often associated with severe side effects. As research advances, our understanding of the cellular and molecular mechanisms underlying the development and maintenance of auto-reactive B cell responses has significantly advanced. As a result, novel immunotherapies aimed to restore immune tolerance and prevent disease progression in autoimmune patients are underway. In this regard, encouraging results from clinical and preclinical studies demonstrate that subcutaneous administration of low-doses of recombinant Interleukin-2 (r-IL2) has potent immunosuppressive effects in patients with autoimmune pathologies. Although the exact mechanism by which IL-2 induces immunosuppression remains unclear, the clinical benefits of the current IL-2-based immunotherapies are attributed to its effect on bolstering T regulatory (Treg) cells, which are known to suppress overactive immune responses. In addition to Tregs, however, rIL-2 also directly prevent the T follicular helper cells (Tfh), T helper 17 cells (Th17), and Double Negative (DN) T cell responses, which play critical roles in the development of autoimmune disorders and have the ability to help pathogenic B cells. Here we discuss the broader effects of rIL-2 immunotherapy and the potential of combining rIL-2 with other cytokine-based therapies to more efficiently target Tfh cells, Th17, and DN T cells and subsequently inhibit auto-antibody (ab) production in autoimmune patients.
Collapse
Affiliation(s)
| | - André Ballesteros-Tato
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW TCRαβ+CD4-CD8- double-negative T (DNT) cells, a principal subset of mature T lymphocytes, have been closely linked with autoimmune/inflammatory conditions. However, controversy persists regarding their ontogeny and function. Here, we present an overview on DNT cells in different autoimmune diseases to advance a deeper understanding of the contribution of this population to disease pathogenesis. RECENT FINDINGS DNT cells have been characterized in various chronic inflammatory diseases and they have been proposed to display pathogenic or regulatory function. The tissue location of DNT cells and the effector cytokines they produce bespeak to their active involvement in chronic inflammatory diseases. SUMMARY By producing various cytokines, expanded DNT cells in inflamed tissues contribute to the pathogenesis of a variety of autoimmune inflammatory diseases. However, it is unclear whether this population represents a stable lineage consisting of different subsets similar to CD4+ T helper cell subset. Better understanding of the possible heterogeneity and plasticity of DNT cells is needed to reveal interventional therapeutic opportunities.
Collapse
Affiliation(s)
- Hao Li
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
19
|
Key diagnostic markers for autoimmune lymphoproliferative syndrome with molecular genetic diagnosis. Blood 2020; 136:1933-1945. [DOI: 10.1182/blood.2020005486] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/29/2020] [Indexed: 01/01/2023] Open
Abstract
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a rare immunodeficiency caused by mutations in genes affecting the extrinsic apoptotic pathway (FAS, FASL, CASP10). This study evaluated the clinical manifestations, laboratory findings, and molecular genetic results of 215 patients referred as possibly having ALPS. Double-negative T-cell (DNT) percentage and in vitro apoptosis functional tests were evaluated by fluorescence-activated cell sorting; interleukin 10 (IL-10) and IL-18 and soluble FAS ligand (sFASL) were measured by enzyme-linked immunosorbent assay. Genetic analysis was performed by next-generation sequencing. Clinical background data were collected from patients’ records. Patients were categorized into definite, suspected, or unlikely ALPS groups, and laboratory parameters were compared among these groups. Of 215 patients, 38 met the criteria for definite ALPS and 17 for suspected ALPS. The definite and suspected ALPS patient populations showed higher DNT percentages than unlikely ALPS and had higher rates of lymphoproliferation. Definite ALPS patients had a significantly more abnormal in vitro apoptosis function, with lower annexin, than patients with suspected ALPS (P = .002) and patients not meeting ALPS criteria (P < .001). The combination of elevated DNTs and an abnormal in vitro apoptosis functional test was the most useful in identifying all types of ALPS patients; the combination of an abnormal in vitro apoptosis functional test and elevated sFASLs was a predictive marker for ALPS-FAS group identification. Lymphoproliferation, apoptosis functional test, and DNTs are the most sensitive markers; elevated IL-10 and IL-18 are additional indicators for ALPS. The combination of elevated sFASLs and abnormal apoptosis function was the most valuable prognosticator for patients with FAS mutations.
Collapse
|
20
|
Alexander JJ, Jacob A, Chang A, Quigg RJ, Jarvis JN. Double negative T cells, a potential biomarker for systemic lupus erythematosus. PRECISION CLINICAL MEDICINE 2020; 3:34-43. [PMID: 32257532 PMCID: PMC7093895 DOI: 10.1093/pcmedi/pbaa001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/17/2019] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that is a challenge to diagnose and treat. There is an urgent need for biomarkers to help define organ involvement, and more effective therapies. A unique population of T cells, the CD3+CD4−CD8− (DNeg) cells, is significantly increased in lupus patients. Twenty-seven cases (53%) of pediatric SLE patients had elevated DNeg cells in their peripheral blood, which correlated with kidney function (R2 = 0.54). Significant infiltration of DNeg cells was observed in both adult and pediatric lupus kidneys by immunofluorescence. For the first time, this study provides direct evidence that DNeg cells facilitate kidney injury in preclinical 8-week-old MRL/lpr lupus mice. In lupus mice, the increase in DNeg cells tracked with worsening disease and correlated with kidney function (R2 = 0.85). Our results show that DNeg cells per se can cause kidney dysfunction, increase in number with increase in disease pathology, and could serve as a potential biomarker.
Collapse
Affiliation(s)
- Jessy J Alexander
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.,Departments of Medicine, Pediatrics, Jacobs School of Medicine and Biomedical Sciences and Genetics, Genomics, & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - Alexander Jacob
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.,Departments of Medicine, Pediatrics, Jacobs School of Medicine and Biomedical Sciences and Genetics, Genomics, & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - Anthony Chang
- Departments of Medicine, Pediatrics, Jacobs School of Medicine and Biomedical Sciences and Genetics, Genomics, & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - Richard J Quigg
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.,Departments of Medicine, Pediatrics, Jacobs School of Medicine and Biomedical Sciences and Genetics, Genomics, & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - James N Jarvis
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
21
|
Liapis K, Tsagarakis NJ, Panitsas F, Taparkou A, Liapis I, Roubakis C, Tsokanas D, Vasileiou P, Grigoriou E, Kakiopoulos G, Psarra K, Farmaki E, Paterakis G. Causes of double-negative T-cell lymphocytosis in children and adults. J Clin Pathol 2019; 73:431-438. [PMID: 31810993 DOI: 10.1136/jclinpath-2019-206255] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 01/18/2023]
Abstract
AIMS The causes and diagnosis of 'double-negative' (CD3+CD4-CD8-) T-cell lymphocytosis are not well studied. We aimed to define the causes of double-negative T-cell lymphocytosis in children and adults, and to identify simple clinical and laboratory features that would help to differentiate between the underlying conditions. METHODS We collected clinical and laboratory data on 10 children and 30 adults with significantly increased peripheral-blood double-negative T-cells (>10% of total lymphocytes). We identified conditions associated with double-negative T-lymphocytosis with flow cytometry, peripheral-blood morphology and T-cell receptor-gene rearrangement studies. Patients were assigned to diagnostic categories on the basis of these test results. RESULTS AND CONCLUSIONS The causes of double-negative T-cell lymphocytosis in children were autoimmune lymphoproliferative syndrome (ALPS) and reactive γ/δ Τ-lymphocytosis. T-cell large granular lymphocyte (T-LGL) leukaemia, reactive γ/δ T-lymphocytosis and hepatosplenic T-cell lymphoma (HSTL) were the the most common disorders underlying double-negative T-cell lymphocytosis in adults. Less common causes included hypereosinophilic syndrome, peripheral T-cell lymphoma, ALPS and monoclonal, double-negative T-lymphocytosis of uncertain significance. CD5/CD7/Vδ2 expression and absolute double-negative lymphocyte count (<1.8×109/L) were useful discriminators for distinguishing patients with reactive γ/δ T-lymphocytosis from those with γ/δ lymphoproliferative disorders. Differentiating between γ/δ T-LGL and HSTL can be difficult. Expression of CD57 and cellular morphology (pale cytoplasm with distinct granules) would support a diagnosis of γ/δ T-LGL.
Collapse
Affiliation(s)
- Konstantinos Liapis
- Department of Clinical Haematology, Peripheral General Hospital Athens Giorgos Gennimatas, Athens, Greece
| | - Nikolaos J Tsagarakis
- Department of Immunology, Peripheral General Hospital Athens Giorgos Gennimatas, Athens, Greece
| | - Fotis Panitsas
- Department of Haematology, General University Hospital of Larisa, Larissa, Thessaly, Greece
| | - Anna Taparkou
- Paediatric Immunology and Rheumatology Referral Center, First Department of Paediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Liapis
- Department of Haematology, University Hospital of Crete, Heraklion, Greece
| | | | - Dimitris Tsokanas
- Department of Clinical Haematology, Peripheral General Hospital Athens Giorgos Gennimatas, Athens, Greece
| | - Paraskevi Vasileiou
- Department of Flow Cytometry, Flowdiagnosis Diagnostic Center, Athens, Greece
| | - Eirini Grigoriou
- Flow Cytometry Laboratory, Department of Immunology and Histocompatibility, Evangelismos Athens General Hospital, Athens, Greece
| | - Georgios Kakiopoulos
- Department of Pathology, Peripheral General Hospital Athens Giorgos Gennimatas, Athens, Greece
| | - Katerina Psarra
- Flow Cytometry Laboratory, Department of Immunology and Histocompatibility, Evangelismos Athens General Hospital, Athens, Greece
| | - Evangelia Farmaki
- Paediatric Immunology and Rheumatology Referral Center, First Department of Paediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George Paterakis
- Department of Immunology, Peripheral General Hospital Athens Giorgos Gennimatas, Athens, Greece
| |
Collapse
|
22
|
Cabral-Marques O, Schimke LF, de Oliveira EB, El Khawanky N, Ramos RN, Al-Ramadi BK, Segundo GRS, Ochs HD, Condino-Neto A. Flow Cytometry Contributions for the Diagnosis and Immunopathological Characterization of Primary Immunodeficiency Diseases With Immune Dysregulation. Front Immunol 2019; 10:2742. [PMID: 31849949 PMCID: PMC6889851 DOI: 10.3389/fimmu.2019.02742] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Almost 70 years after establishing the concept of primary immunodeficiency disorders (PIDs), more than 320 monogenic inborn errors of immunity have been identified thanks to the remarkable contribution of high-throughput genetic screening in the last decade. Approximately 40 of these PIDs present with autoimmune or auto-inflammatory symptoms as the primary clinical manifestation instead of infections. These PIDs are now recognized as diseases of immune dysregulation. Loss-of function mutations in genes such as FOXP3, CD25, LRBA, IL-10, IL10RA, and IL10RB, as well as heterozygous gain-of-function mutations in JAK1 and STAT3 have been reported as causative of these disorders. Identifying these syndromes has considerably contributed to expanding our knowledge on the mechanisms of immune regulation and tolerance. Although whole exome and whole genome sequencing have been extremely useful in identifying novel causative genes underlying new phenotypes, these approaches are time-consuming and expensive. Patients with monogenic syndromes associated with autoimmunity require faster diagnostic tools to delineate therapeutic strategies and avoid organ damage. Since these PIDs present with severe life-threatening phenotypes, the need for a precise diagnosis in order to initiate appropriate patient management is necessary. More traditional approaches such as flow cytometry are therefore a valid option. Here, we review the application of flow cytometry and discuss the relevance of this powerful technique in diagnosing patients with PIDs presenting with immune dysregulation. In addition, flow cytometry represents a fast, robust, and sensitive approach that efficiently uncovers new immunopathological mechanisms underlying monogenic PIDs.
Collapse
Affiliation(s)
- Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | | | - Nadia El Khawanky
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg im Breisgau, Germany.,Precision Medicine Theme, The South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Rodrigo Nalio Ramos
- INSERM U932, SiRIC Translational Immunotherapy Team, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Basel K Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | | | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, WA, United States
| | - Antonio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
23
|
Cagdas D, Halaçlı SO, Tan Ç, Lo B, Çetinkaya PG, Esenboğa S, Karaatmaca B, Matthews H, Balcı-Hayta B, Arıkoğlu T, Ezgü F, Aladağ E, Saltık-Temizel İN, Demir H, Kuşkonmaz B, Okur V, Gümrük F, Göker H, Çetinkaya D, Boztuğ K, Lenardo M, Sanal Ö, Tezcan İ. A Spectrum of Clinical Findings from ALPS to CVID: Several Novel LRBA Defects. J Clin Immunol 2019; 39:726-738. [PMID: 31432443 PMCID: PMC11090043 DOI: 10.1007/s10875-019-00677-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/25/2019] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Autosomal recessively inherited lipopolysaccharide-responsive beige-like anchor (LRBA) protein deficiency was shown to be responsible for different types of inborn errors of immunity, such as common variable immunodeficiency (CVID) and autoimmune lymphoproliferative syndrome (ALPS). The aim of this study was to compare patients with LRBA-related ALPS and LRBA-related CVID, to describe their clinical and laboratory phenotypes, and to prepare an algorithm for their diagnosis and management. METHODS Fifteen LRBA-deficient patients were identified among 31 CVID and 14 possible ALPS patients with Western blotting (WB), primary immunodeficiency disease (PIDD) gene, next-generation panel screening (NGS), and whole exome sequencing (WES). RESULTS The median age on admission and age of diagnosis were 7 years (0.3-16.5) and 11 years (5-44), respectively. Splenomegaly was seen in 93.3% (14/15) of the patients on admission. Splenectomy was performed to 1/5. Recurrent upper respiratory tract infections (93.3% (14/15)), autoimmune cytopenia (80% (12/15)), chronic diarrhea (53.3% (8/15)), lower respiratory tract infections (53.3% (8/15)), lymphoma (26.6% (4/15)), Evans syndrome (26.6% (4/15)), and autoimmune thyroiditis (20% (3/15)) were common clinical findings and diseases. Lymphopenia (5/15), intermittant neutropenia (4/15), eosinophilia (4/15), and progressive hypogammaglobulinemia are recorded in given number of patients. Double negative T cells (TCRαβ+CD4-CD8-) were increased in 80% (8/10) of the patients. B cell percentage/numbers were low in 60% (9/15) of the patients on admission. Decreased switched memory B cells, decreased naive and recent thymic emigrant (RTE) Thelper (Th) cells, markedly increased effector memory/effector memory RA+ (TEMRA) Th were documented. Large PD1+ population, increased memory, and enlarged follicular helper T cell population in the CD4+ T cell compartment was seen in one of the patients. Most of the deleterious missense mutations were located in the DUF1088 and BEACH domains. Interestingly, one of the two siblings with the same homozygous LRBA defect did not have any clinical symptom. Hematopoietic stem cell transplantation (HSCT) was performed to 7/15 (46.6%) of the patients. Transplanted patients are alive and well after a median of 2 years (1-3). In total, one patient died from sepsis during adulthood before HSCT. CONCLUSION Patients with LRBA deficiency may initially be diagnosed as CVID or ALPS in the clinical practice. Progressive decrease in B cells as well as IgG in ALPS-like patients and addition of IBD symptoms in the follow-up should raise the suspicion for LRBA deficiency. Decreased switched memory B cells, decreased naive and recent thymic emigrant (RTE) Th cells, and markedly increased effector memory/effector memory RA+ Th cells (TEMRA Th) cells are important for the diagnosis of the patients in addition to clinical features. Analysis of protein by either WB or flow cytometry is required when the clinicians come across especially with missense LRBA variants of uncertain significance. High rate of malignancy shows the regulatory T cell's important role of immune surveillance. HSCT is curative and succesful in patients with HLA-matched family donor.
Collapse
Affiliation(s)
- Deniz Cagdas
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University Medical School, Ankara, Turkey.
| | | | - Çağman Tan
- Institute of Child Health, Immunology, Hacettepe University, Ankara, Turkey
| | - Bernice Lo
- Sidra Medical and Research Center, Al Rayyan, Qatar
| | - Pınar Gür Çetinkaya
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University Medical School, Ankara, Turkey
| | - Saliha Esenboğa
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University Medical School, Ankara, Turkey
| | - Betül Karaatmaca
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University Medical School, Ankara, Turkey
| | - Helen Matthews
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Burcu Balcı-Hayta
- Department of Medical Biology, Hacettepe University Medical School, Ankara, Turkey
| | - Tuba Arıkoğlu
- Department of Pediatrics, Division of Allergy and Immunology, Mersin University Medical School, Mersin, Turkey
| | - Fatih Ezgü
- Department of Pediatrics, Division of Pediatric Inborn Metabolic Disorders, Metabolism and Genetics, Gazi University Medical School, Ankara, Turkey
| | - Elifcan Aladağ
- Department of Internal Medicine, Division of Hematology, Hacettepe University Medical School, Ankara, Turkey
| | - İnci N Saltık-Temizel
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hacettepe University Medical School, Ankara, Turkey
| | - Hülya Demir
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hacettepe University Medical School, Ankara, Turkey
| | - Barış Kuşkonmaz
- Department of Pediatrics, Division of Pediatric Hematology, Hacettepe University Medical School, Ankara, Turkey
| | - Visal Okur
- Department of Pediatrics, Division of Pediatric Hematology, Hacettepe University Medical School, Ankara, Turkey
| | - Fatma Gümrük
- Department of Pediatrics, Division of Pediatric Hematology, Hacettepe University Medical School, Ankara, Turkey
| | - Hakan Göker
- Department of Internal Medicine, Division of Hematology, Hacettepe University Medical School, Ankara, Turkey
| | - Duygu Çetinkaya
- Department of Pediatrics, Division of Pediatric Hematology, Hacettepe University Medical School, Ankara, Turkey
| | - Kaan Boztuğ
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Michael Lenardo
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Özden Sanal
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University Medical School, Ankara, Turkey
| | - İlhan Tezcan
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University Medical School, Ankara, Turkey
| |
Collapse
|
24
|
Tay SH, Yaung KN, Leong JY, Yeo JG, Arkachaisri T, Albani S. Immunomics in Pediatric Rheumatic Diseases. Front Med (Lausanne) 2019; 6:111. [PMID: 31231652 PMCID: PMC6558393 DOI: 10.3389/fmed.2019.00111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/03/2019] [Indexed: 02/04/2023] Open
Abstract
The inherent complexity in the immune landscape of pediatric rheumatic disease necessitates a holistic system approach. Uncertainty in the mechanistic workings and etiological driving forces presents difficulty in personalized treatments. The development and progression of immunomics are well suited to deal with this complexity. Immunomics encompasses a spectrum of biological processes that entail genomics, transcriptomics, epigenomics, proteomics, and cytomics. In this review, we will discuss how various high dimensional technologies in immunomics have helped to grow a wealth of data that provide salient clues and biological insights into the pathogenesis of autoimmunity. Interfaced with critical unresolved clinical questions and unmet medical needs, these platforms have helped to identify candidate immune targets, refine patient stratification, and understand treatment response or resistance. Yet the unprecedented growth in data has presented both opportunities and challenges. Researchers are now facing huge heterogeneous data sets from different origins that need to be integrated and exploited for further data mining. We believe that the utilization and integration of these platforms will help unravel the complexities and expedite both discovery and validation of clinical targets.
Collapse
Affiliation(s)
| | | | - Jing Yao Leong
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Joo Guan Yeo
- Duke-NUS Medical School, Singapore, Singapore.,Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.,Rheumatology and Immunology Service, Department of Pediatric Subspecialties, KK Women's and Children's Hospital, Singapore, Singapore
| | - Thaschawee Arkachaisri
- Duke-NUS Medical School, Singapore, Singapore.,Rheumatology and Immunology Service, Department of Pediatric Subspecialties, KK Women's and Children's Hospital, Singapore, Singapore
| | - Salvatore Albani
- Duke-NUS Medical School, Singapore, Singapore.,Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.,Rheumatology and Immunology Service, Department of Pediatric Subspecialties, KK Women's and Children's Hospital, Singapore, Singapore
| |
Collapse
|
25
|
Wang Y, Lu W, Li A, Sun Z, Wang L. Elevated CD3 low double negative T lymphocyte is associated with pneumonia and its severity in pediatric patients. PeerJ 2018; 6:e6114. [PMID: 30588404 PMCID: PMC6302782 DOI: 10.7717/peerj.6114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 11/15/2018] [Indexed: 11/28/2022] Open
Abstract
Background Previous studies have shown that the adaptive immunity function of T cells in disease states correlates with CD3 surface expression closely. During routine assessment of TBNK subsets in peripheral blood of pediatric patients by flow cytometry, we noticed that variable expression levels of CD3 on CD3+CD4−CD8− double-negative T (DNT) lymphocytes in different patients. The objective of this study was to assess the relationship of CD3 expression levels on DNT cells with disease severity. Methods In this prospective study, we investigated the frequencies of circulating CD4−CD8− DNT cell subsets with CD3low or CD3high phenotype by flow cytometry in 76 pediatric patients with pneumonia, 55 patients with severe pneumonia (SP), and 29 healthy controls (Con). Results The numbers of circulating DNT cells were similar in all groups; however, the frequency of CD3low DNT cell subsets was significantly increased in patients with pneumonia (p < 0.001) and SP (p < 0.001). The elevated CD3low DNT cell frequency showed a positive correlation with the clinical severity of pneumonia. On sub-group analysis, the frequency of CD3low DNT cells was only elevated in children with pneumonia aged <5 years, while no association was observed with the causative pathogen of pneumonia. Conclusions These findings suggest that CD3 expression levels on DNT cell subsets of peripheral lymphocytes may be a valuable biomarker for evaluation of immune response in pediatric infectious disease. CD3low DNT cells were elevated in children with pneumonia aged <5 years, which indicates that it may be an important research target in pediatric infectious diseases.
Collapse
Affiliation(s)
- Ying Wang
- Institute of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Wenting Lu
- Department of Molecular Biology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Aipeng Li
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, China
| | - Zhengyi Sun
- Institute of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Liying Wang
- Institute of Pediatrics, The First Hospital of Jilin University, Changchun, China.,Department of Molecular Biology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| |
Collapse
|
26
|
Brandt D, Hedrich CM. TCRαβ +CD3 +CD4 -CD8 - (double negative) T cells in autoimmunity. Autoimmun Rev 2018; 17:422-430. [PMID: 29428806 DOI: 10.1016/j.autrev.2018.02.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 12/07/2017] [Indexed: 12/14/2022]
Abstract
TCRαβ+CD3+CD4-CD8- "double negative" (DN) T cells comprise a small subset of mature peripheral T cells. The origin and function of DN T cells are somewhat unclear and discussed controversially. While DN T cells resemble a rare and heterogeneous T cell subpopulation in healthy individuals, numbers of TCRαβ+ DN T cells are expanded in several inflammatory conditions, where they also exhibit distinct effector phenotypes and infiltrate inflamed tissues. Thus, DN T cells may be involved in systemic inflammation and tissue damage in autoimmune/inflammatory conditions, including SLE, Sjögren's syndrome, and psoriasis. Here, the current understanding of the origin and phenotype of DN T cells, and their role in the instruction of immune responses, autoimmunity and inflammation will be discussed in health and disease.
Collapse
Affiliation(s)
- D Brandt
- Division of Pediatric Rheumatology and Immunology, Children's Hospital Dresden, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - C M Hedrich
- Division of Pediatric Rheumatology and Immunology, Children's Hospital Dresden, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany; Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK.
| |
Collapse
|
27
|
Chen X, Kunda PE, Lin J, Zhou M, Huang J, Zhang H, Liu T. SYK-targeted dendritic cell-mediated cytotoxic T lymphocytes enhance the effect of immunotherapy on retinoblastoma. J Cancer Res Clin Oncol 2018; 144:675-684. [PMID: 29372378 PMCID: PMC5843685 DOI: 10.1007/s00432-018-2584-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022]
Abstract
PURPOSE Retinoblastoma (RB) is the most common primary intraocular tumor in children. Chemotherapy is currently the main method of RB treatment. Unfortunately, RB often becomes chemoresistant and turns lethal. Here, we used in vitro cell immunotherapy to explore whether adoptive immunotherapy could be used as a potential treatment for RB. We focused on spleen tyrosine kinase (SYK), which is significantly upregulated in RB cells and serves as a marker for RB cells. METHODS Using lentiviruses, we genetically modified dendritic cells (DCs) to express and present the SYK peptide antigen to cytotoxic T lymphocytes (CTLs) in vitro. We used SYK-negative cell lines (MDA-MB-231, MCF-10A, and hTERT-RPE1) and SYK-positive cell lines (MCF-7 and RB-Y79) to evaluate the specificity and cytotoxicity of DC presented CTLs using FACS, live-cell imaging, and RNA interference. RESULTS The cytotoxicity of CTLs induced by SYK-overexpressing DCs (SYK-DC-CTLs) was enhanced more than three times in SYK-positive cell lines compared with SYK-negative cell lines. DCs primed with SYK could drive CTL cytotoxicity against SYK-positive cell lines but not against SYK-negative cell lines. Moreover, SYK-silenced RB-Y79 cells successfully evaded the cytotoxic attack from SYK-DC-CTLs. However, SYK-DC-CTLs could target SYK overexpressed hTERT-RPE1 cells, suggesting that SYK is a specific antigen for RB. Furthermore, SYK-DC-CTL exhibited specific cytotoxicity against carboplatin-resistant RB-Y79 cells in vitro. CONCLUSIONS Our data showed that SYK could be a potential immunotherapy target mediated by DCs. We propose SYK as a candidate target for treatment of chemoresistant RB.
Collapse
Affiliation(s)
- Xuemei Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Patricia Elena Kunda
- Centro Investigación Medicina Traslacional "Severo Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Córdoba, Argentina
| | - Jianwei Lin
- Shenzhen Key Laboratory for Anti-Ageing and Regenerative Medicine, Health Science Center, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, Guangdong, China
| | - Meiling Zhou
- Shenzhen Key Laboratory for Anti-Ageing and Regenerative Medicine, Health Science Center, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, Guangdong, China
- Department of Biotherapy, Shenzhen Luohu People's Hospital, No. 47 Youyi Road, Shenzhen, 518001, Guangdong, China
| | - Jinghan Huang
- Department of Biotherapy, Shenzhen Luohu People's Hospital, No. 47 Youyi Road, Shenzhen, 518001, Guangdong, China
| | - Huqin Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Tao Liu
- Shenzhen Key Laboratory for Anti-Ageing and Regenerative Medicine, Health Science Center, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, Guangdong, China.
- Department of Biotherapy, Shenzhen Luohu People's Hospital, No. 47 Youyi Road, Shenzhen, 518001, Guangdong, China.
| |
Collapse
|
28
|
Signal transducer and activator of transcription gain-of-function primary immunodeficiency/immunodysregulation disorders. Curr Opin Pediatr 2017; 29:711-717. [PMID: 28914637 DOI: 10.1097/mop.0000000000000551] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW To describe primary immunodeficiencies caused by gain-of-function (GOF) mutations of signal transducer and activator of transcription (STAT) genes, a group of genetically determined disorders characterized by susceptibility to infections and, in many cases, autoimmune manifestations. RECENT FINDINGS GOF mutations affecting STAT1 result in increased STAT tyrosine phosphorylation and secondarily increased response to STAT1-signaling cytokines, such as interferons. In contrast, STAT3 hyperactivity is not usually related to hyperphosphorylation but rather to increased STAT3-mediated transcriptional activity. In both cases, heterozygous STAT1 and STAT3 GOF mutations trigger a distinct set of genes in target cells that lead to abnormal functioning of antimicrobial response and/or autoimmunity and result in autosomal dominant diseases. SUMMARY Clinical manifestations of patients with STAT1 GOF are characterized by mucocutaneous candidiasis and recurrent lower tract respiratory infections. In addition, many patients have thyroiditis, type 1 diabetes mellitus, autoimmune cytopenias, cancer or aneurysms. Patients with germline STAT3 GOF mutations have an increased frequency of early-onset multiorgan autoimmunity (i.e. autoimmune enteropathy, type 1 diabetes mellitus, autoimmune interstitial lung disease and autoimmune cytopenias), lymphoproliferation, short stature and, less frequently, severe recurrent infections. Treatment options range from antimicrobial therapy, intravenous or subcutaneous immunoglobulin and immunosuppressive drugs. Some patients with STAT1 GOF disorder have undergone hematopoietic stem cell transplantation, although these have been difficult because of the underlying proinflammatory milieu from the mutation.
Collapse
|
29
|
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is an inherited syndrome characterized by abnormal lymphocyte survival caused by failure of apoptotic mechanisms to maintain lymphocyte homeostasis. This failure leads to the clinical manifestations of non-infectious and non-malignant lymphadenopathy, splenomegaly, and autoimmune pathology, most commonly, autoimmune cytopenias. Since ALPS was first characterized in the early 1990s, insights in disease biology have improved both diagnosis and management of this syndrome. Sirolimus is the best-studied and most effective corticosteroid-sparing therapy for ALPS and should be considered first-line for patients in need of chronic treatment. This review highlights practical clinical considerations for the diagnosis and management of ALPS. Further studies could reveal new proteins and regulatory pathways that are critical for lymphocyte activation and apoptosis.
Collapse
Affiliation(s)
- Karen Bride
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - David Teachey
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
30
|
El-Sayed ZA, El-Owaidy RH, Mohamed NL, Shehata BA. Alpha beta double negative T cells in children with systemic lupus erythematosus: The relation to disease activity and characteristics. Mod Rheumatol 2017; 28:654-660. [DOI: 10.1080/14397595.2017.1377146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Zeinab A. El-Sayed
- Pediatric Allergy and Immunology Unit, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Rasha H. El-Owaidy
- Pediatric Allergy and Immunology Unit, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Neama L. Mohamed
- Clinical Pathology Department, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Beshoy A. Shehata
- Pediatric Allergy and Immunology Unit, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| |
Collapse
|
31
|
|
32
|
Abnormal hematopoiesis and autoimmunity in human subjects with germline IKZF1 mutations. J Allergy Clin Immunol 2017; 140:223-231. [DOI: 10.1016/j.jaci.2016.09.029] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/08/2016] [Accepted: 09/23/2016] [Indexed: 01/28/2023]
|
33
|
Agrebi N, Ben-Mustapha I, Matoussi N, Dhouib N, Ben-Ali M, Mekki N, Ben-Ahmed M, Larguèche B, Ben Becher S, Béjaoui M, Barbouche MR. Rare splicing defects of FAS underly severe recessive autoimmune lymphoproliferative syndrome. Clin Immunol 2017; 183:17-23. [PMID: 28668589 DOI: 10.1016/j.clim.2017.06.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 06/08/2017] [Accepted: 06/25/2017] [Indexed: 10/19/2022]
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a prototypic disorder of impaired apoptosis characterized by autoimmune features and lymphoproliferation. Heterozygous germline or somatic FAS mutations associated with preserved protein expression have been described. Very rare cases of homozygous germline FAS mutations causing severe autosomal recessive form of ALPS with a complete defect of Fas expression have been reported. We report two unrelated patients from highly inbred North African population showing a severe ALPS phenotype and an undetectable Fas surface expression. Two novel homozygous mutations have been identified underlying rare splicing defects mechanisms. The first mutation breaks a branch point sequence and the second alters a regulatory exonic splicing site. These splicing defects induce the skipping of exon 6 encoding the transmembrane domain of CD95. Our findings highlight the requirement of tight regulation of FAS exon 6 splicing for balanced alternative splicing and illustrate the importance of such studies in highly consanguineous populations.
Collapse
Affiliation(s)
- N Agrebi
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, 1002 Tunis, Belvédère, Tunisia; Université de Tunis El Manar, 1068 Tunis, Tunisia; The University of Carthage, Faculty of Sciences of Bizerte, 7021 Jarzouna, Tunisia
| | - I Ben-Mustapha
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, 1002 Tunis, Belvédère, Tunisia; Université de Tunis El Manar, 1068 Tunis, Tunisia; Faculty of Medicine, 1007 Tunis, Tunisia.
| | - N Matoussi
- Faculty of Medicine, 1007 Tunis, Tunisia; Department of Pediatric Care, Emergency and Out Patient Children's Hospital of Tunis, 1029 Tunis, Tunisia
| | - N Dhouib
- Faculty of Medicine, 1007 Tunis, Tunisia; National Bone Marrow Transplantation Center, 1006 Tunis, Tunisia
| | - M Ben-Ali
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, 1002 Tunis, Belvédère, Tunisia; Université de Tunis El Manar, 1068 Tunis, Tunisia
| | - N Mekki
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, 1002 Tunis, Belvédère, Tunisia; Université de Tunis El Manar, 1068 Tunis, Tunisia; Faculty of Medicine, 1007 Tunis, Tunisia
| | - M Ben-Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, 1002 Tunis, Belvédère, Tunisia; Université de Tunis El Manar, 1068 Tunis, Tunisia; Faculty of Medicine, 1007 Tunis, Tunisia
| | - B Larguèche
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, 1002 Tunis, Belvédère, Tunisia
| | - S Ben Becher
- Faculty of Medicine, 1007 Tunis, Tunisia; Department of Pediatric Care, Emergency and Out Patient Children's Hospital of Tunis, 1029 Tunis, Tunisia
| | - M Béjaoui
- Faculty of Medicine, 1007 Tunis, Tunisia; National Bone Marrow Transplantation Center, 1006 Tunis, Tunisia
| | - M R Barbouche
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, 1002 Tunis, Belvédère, Tunisia; Université de Tunis El Manar, 1068 Tunis, Tunisia; Faculty of Medicine, 1007 Tunis, Tunisia
| |
Collapse
|
34
|
Celhar T, Fairhurst AM. Modelling clinical systemic lupus erythematosus: similarities, differences and success stories. Rheumatology (Oxford) 2017; 56:i88-i99. [PMID: 28013204 PMCID: PMC5410990 DOI: 10.1093/rheumatology/kew400] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Indexed: 12/26/2022] Open
Abstract
Mouse models of SLE have been indispensable tools to study disease pathogenesis, to identify genetic susceptibility loci and targets for drug development, and for preclinical testing of novel therapeutics. Recent insights into immunological mechanisms of disease progression have boosted a revival in SLE drug development. Despite promising results in mouse studies, many novel drugs have failed to meet clinical end points. This is probably because of the complexity of the disease, which is driven by polygenic predisposition and diverse environmental factors, resulting in a heterogeneous clinical presentation. Each mouse model recapitulates limited aspects of lupus, especially in terms of the mechanism underlying disease progression. The main mouse models have been fairly successful for the evaluation of broad-acting immunosuppressants. However, the advent of targeted therapeutics calls for a selection of the most appropriate model(s) for testing and, ultimately, identification of patients who will be most likely to respond.
Collapse
Affiliation(s)
- Teja Celhar
- Singapore Immunology Network, A*STAR, Singapore, Republic of Singapore
| | - Anna-Marie Fairhurst
- Singapore Immunology Network, A*STAR, Singapore, Republic of Singapore.,Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
35
|
Nabhani S, Schipp C, Miskin H, Levin C, Postovsky S, Dujovny T, Koren A, Harlev D, Bis AM, Auer F, Keller B, Warnatz K, Gombert M, Ginzel S, Borkhardt A, Stepensky P, Fischer U. STAT3 gain-of-function mutations associated with autoimmune lymphoproliferative syndrome like disease deregulate lymphocyte apoptosis and can be targeted by BH3 mimetic compounds. Clin Immunol 2017; 181:32-42. [PMID: 28579554 DOI: 10.1016/j.clim.2017.05.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 05/19/2017] [Accepted: 05/31/2017] [Indexed: 01/19/2023]
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is typically caused by mutations in genes of the extrinsic FAS mediated apoptotic pathway, but for about 30% of ALPS-like patients the genetic diagnosis is lacking. We analyzed 30 children with ALPS-like disease of unknown cause and identified two dominant gain-of-function mutations of the Signal Transducer And Activator Of Transcription 3 (STAT3, p.R278H, p.M394T) leading to increased transcriptional activity. Hyperactivity of STAT3, a known repressor of FAS, was associated with decreased FAS-mediated apoptosis, mimicking ALPS caused by FAS mutations. Expression of BCL2 family proteins, further targets of STAT3 and regulators of the intrinsic apoptotic pathway, was disturbed. Cells with hyperactive STAT3 were consequently more resistant to intrinsic apoptotic stimuli and STAT3 inhibition alleviated this effect. Importantly, STAT3-mutant cells were more sensitive to death induced by the BCL2-inhibitor ABT-737 indicating a dependence on anti-apoptotic BCL2 proteins and potential novel therapeutic options.
Collapse
Affiliation(s)
- Schafiq Nabhani
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Cyrill Schipp
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Hagit Miskin
- Pediatric Hematology Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Carina Levin
- Pediatric Hematology Unit, Emek Medical Center, Afula, Israel
| | - Sergey Postovsky
- Department of Pediatric Oncology/Hematology Meyer Children's Hospital Rambam Health Care, Haifa, Israel
| | - Tal Dujovny
- Pediatric Hematology Unit, Emek Medical Center, Afula, Israel
| | - Ariel Koren
- Pediatric Hematology Unit, Emek Medical Center, Afula, Israel
| | - Dan Harlev
- Pediatric Hematology Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Anne-Marie Bis
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Franziska Auer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Baerbel Keller
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Klaus Warnatz
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Michael Gombert
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Sebastian Ginzel
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Department of Computer Science, Bonn-Rhine-Sieg University of Applied Sciences, Sankt Augustin, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Polina Stepensky
- Department of Bone Marrow Transplantation, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany.
| |
Collapse
|
36
|
Chowdhary VR, Krogman A, Tilahun AY, Alexander MP, David CS, Rajagopalan G. Concomitant Disruption of CD4 and CD8 Genes Facilitates the Development of Double Negative αβ TCR + Peripheral T Cells That Respond Robustly to Staphylococcal Superantigen. THE JOURNAL OF IMMUNOLOGY 2017; 198:4413-4424. [PMID: 28468970 DOI: 10.4049/jimmunol.1601991] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/06/2017] [Indexed: 01/14/2023]
Abstract
Mature peripheral double negative T (DNT) cells expressing αβ TCR but lacking CD4/CD8 coreceptors play protective as well as pathogenic roles. To better understand their development and functioning in vivo, we concomitantly inactivated CD4 and CD8 genes in mice with intact MHC class I and class II molecules with the hypothesis that this would enable the development of DNT cells. We also envisaged that these DNT cells could be activated by bacterial superantigens in vivo as activation of T cells by superantigens does not require CD4 and CD8 coreceptors. Because HLA class II molecules present superantigens more efficiently than murine MHC class II molecules, CD4 CD8 double knockout (DKO) mice transgenically expressing HLA-DR3 or HLA-DQ8 molecules were generated. Although thymic cellularity was comparable between wild type (WT) and DKO mice, CD3+ αβ TCR+ thymocytes were significantly reduced in DKO mice, implying defects in thymic-positive selection. Splenic CD3+ αβ TCR+ cells and Foxp3+ T regulatory cells were present in DKO mice but significantly reduced. However, the in vivo inflammatory responses and immunopathology elicited by acute challenge with the staphylococcal superantigen enterotoxin B were comparable between WT and DKO mice. Choric exposure to staphylococcal enterotoxin B precipitated a lupus-like inflammatory disease with characteristic lympho-monocytic infiltration in lungs, livers, and kidneys, along with production of anti-nuclear Abs in DKO mice as in WT mice. Overall, our results suggest that DNT cells can develop efficiently in vivo and chronic exposure to bacterial superantigens may precipitate a lupus-like autoimmune disease through activation of DNT cells.
Collapse
Affiliation(s)
- Vaidehi R Chowdhary
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Ashton Krogman
- Department of Immunology, Mayo Clinic, Rochester, MN 55905; and
| | | | - Mariam P Alexander
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905
| | - Chella S David
- Department of Immunology, Mayo Clinic, Rochester, MN 55905; and
| | | |
Collapse
|
37
|
Suárez-Fueyo A, Bradley SJ, Klatzmann D, Tsokos GC. T cells and autoimmune kidney disease. Nat Rev Nephrol 2017; 13:329-343. [PMID: 28287110 DOI: 10.1038/nrneph.2017.34] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glomerulonephritis is traditionally considered to result from the invasion of the kidney by autoantibodies and immune complexes from the circulation or following their formation in situ, and by cells of the innate and the adaptive immune system. The inflammatory response leads to the proliferation and dysfunction of cells of the glomerulus, and invasion of the interstitial space with immune cells, resulting in tubular cell malfunction and fibrosis. T cells are critical drivers of autoimmunity and related organ damage, by supporting B-cell differentiation and antibody production or by directly promoting inflammation and cytotoxicity against kidney resident cells. T cells might become activated by autoantigens in the periphery and become polarized to secrete inflammatory cytokines before entering the kidney where they have the opportunity to expand owing to the presence of costimulatory molecules and activating cytokines. Alternatively, naive T cells could enter the kidney where they become activated after encountering autoantigen and expand locally. As not all individuals with a peripheral autoimmune response to kidney antigens develop glomerulonephritis, the contribution of local kidney factors expressed or produced by kidney cells is probably of crucial importance. Improved understanding of the biochemistry and molecular biology of T cells in patients with glomerulonephritis offers unique opportunities for the recognition of treatment targets for autoimmune kidney disease.
Collapse
Affiliation(s)
- Abel Suárez-Fueyo
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, CLS-937, Boston, Massachusetts 02215, USA
| | - Sean J Bradley
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, CLS-937, Boston, Massachusetts 02215, USA
| | - David Klatzmann
- Sorbonne Universités, Pierre and Marie Curie University, INSERM UMR_S 959, 83 Boulevard de l'Hôpital, F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Clinical Investigation Center in Biotherapy and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), 83 boulevard de l'Hôpital, F-75013, Paris, France
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, CLS-937, Boston, Massachusetts 02215, USA
| |
Collapse
|
38
|
Gratzinger D, Jaffe ES, Chadburn A, Chan JKC, de Jong D, Goodlad JR, Said J, Natkunam Y. Primary/Congenital Immunodeficiency: 2015 SH/EAHP Workshop Report-Part 5. Am J Clin Pathol 2017; 147:204-216. [PMID: 28395106 PMCID: PMC6248572 DOI: 10.1093/ajcp/aqw215] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES The 2015 Workshop of the Society for Hematopathology/European Association for Haematopathology aimed to review primary immunodeficiency and related lymphoproliferations. METHODS Primary immunodeficiencies were divided into immune dysregulation, DNA repair defects, low immunoglobulins, and combined immunodeficiencies. RESULTS Autoimmune lymphoproliferative syndrome (ALPS) is a prototypical immune dysregulation-type immunodeficiency, with defects in T-cell signaling or apoptosis, expansion of T-cell subsets, and predisposition to hemophagocytic lymphohistiocytosis. DNA repair defects directly predispose to malignancy. Low immunoglobulin immunodeficiencies such as common variable immunodeficiency (CVID) have underlying T-cell repertoire abnormalities predisposing to autoimmunity and B-cell lymphoproliferations. The full spectrum of B-cell lymphoproliferative disorders occurs in primary immunodeficiency. CONCLUSIONS Lymphoproliferations in primary immunodeficiency mirror those in other immunodeficiency settings, with monomorphic B- and sometimes T lymphoproliferative disorders enriched in DNA repair defects. Distinctive T-cell subset expansions in ALPS, CVID, and related entities can mimic lymphoma, and recognition of double-negative T-cell or cytotoxic T-cell expansions is key to avoid overdiagnosis.
Collapse
Affiliation(s)
- Dita Gratzinger
- From the Stanford University School of Medicine, Stanford, CA
| | | | - Amy Chadburn
- Weill Medical College of Cornell University, New York, NY
| | | | - Daphne de Jong
- VU University Medical Center, Amsterdam, the Netherlands
| | | | - Jonathan Said
- University of California Los Angeles Medical Center, Los Angeles
| | | |
Collapse
|
39
|
Rubio CA, Ichiya T, Schmidt PT. Lymphocytic oesophagitis, eosinophilic oesophagitis and compound lymphocytic-eosinophilic oesophagitis I: histological and immunohistochemical findings. J Clin Pathol 2016; 70:208-216. [PMID: 27471274 DOI: 10.1136/jclinpath-2016-203782] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/21/2016] [Accepted: 07/02/2016] [Indexed: 01/18/2023]
Abstract
AIMS To report four histological-immunohistochemical oesophagitis phenotypes. METHODS Oesophageal biopsies from 311 patients were stained with H&E and with CD3, a T cell marker. Additional immunohistochemical stains (n=413) were performed in 77 cases. RESULTS Four histological-immunohistochemical oesophagitis phenotypes were recorded: lymphocytic oesophagitis (LyE, ≥40 CD3+ lymphocytes/HPF in CD3 immunostain), eosinophilic oesophagitis (EoE, ≥15 eosinophils/HPF in H&E stain), lymphocytic infiltration (≤39 CD3+/HPF) and compound lymphocytic oesophagitis-eosinophilic oesophagitis (Co LyE-EoE). At index biopsy, 28.3% (n=88) had LyE, 21.2% (n=66) EoE, 10.6% (n=33) Co LyE-EoE and 39.9% (n=124) lymphocytic infiltration. A persistent oesophagitis phenotype was found in 42.5% (37/87) in the first follow-up biopsy, in 34.4% (21/61) in the second follow-up biopsy and in 48.1% (26/54) in the third follow-up biopsy. Using βF1 immunostain, two different surface T cell receptors were detected in LyE and Co Lye-EoE: one having ≥40 βF1+/HPF (βF1+ high) and the other having <39 βF1+/HPF (βF1+ low). CONCLUSIONS Based on the literature regarding the significance of intraepithelial lymphocytes (IELs) in the initiation of EoE, we submit that the IEL phenotypes in LyE might differ from those found in EoE as they were unable to elicit the same eosinophilic response. Recent studies disclosed that group 2 innate lymphocytes (ILC2s), enriched in EoE, remain undetected in CD3 immunostain as they lack surface markers for T, B, natural killer (NK) or NK T cells. If ILC2s also participate in the lymphocytic infiltration of EoE, then the frequency of cases with Co LyE-EoE here reported might have been much higher. The four oesophagitis phenotypes described are easy to recognise, provided that the dual staining procedure (H&E-CD3) is implemented.
Collapse
Affiliation(s)
- C A Rubio
- Department of Pathology, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - T Ichiya
- Department of Medicine, Center for Digestive Diseases, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - P T Schmidt
- Department of Medicine, Center for Digestive Diseases, Karolinska Institute and University Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Bride KL, Vincent T, Smith-Whitley K, Lambert MP, Bleesing JJ, Seif AE, Manno CS, Casper J, Grupp SA, Teachey DT. Sirolimus is effective in relapsed/refractory autoimmune cytopenias: results of a prospective multi-institutional trial. Blood 2016; 127:17-28. [PMID: 26504182 PMCID: PMC4705607 DOI: 10.1182/blood-2015-07-657981] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/22/2015] [Indexed: 12/30/2022] Open
Abstract
Patients with autoimmune multilineage cytopenias are often refractory to standard therapies requiring chronic immunosuppression with medications with limited efficacy and high toxicity. We present data on 30 patients treated on a multicenter prospective clinical trial using sirolimus as monotherapy. All children (N = 12) with autoimmune lymphoproliferative syndrome (ALPS) achieved a durable complete response (CR), including rapid improvement in autoimmune disease, lymphadenopathy, and splenomegaly within 1 to 3 months of starting sirolimus. Double-negative T cells were no longer detectable in most, yet other lymphocyte populations were spared, suggesting a targeted effect of sirolimus. We also treated 12 patients with multilineage cytopenias secondary to common variable immunodeficiency (CVID), Evans syndrome (ES), or systemic lupus erythematosus (SLE), and most achieved a CR (N = 8), although the time to CR was often slower than was seen in ALPS. Six children with single-lineage autoimmune cytopenias were treated and only 2 responded. Sirolimus was well tolerated with very few side effects. All of the responding patients have remained on therapy for over 1 year (median, 2 years; range, 1 to 4.5 years). In summary, sirolimus led to CR and durable responses in a majority of children with refractory multilineage autoimmune cytopenias. The responses seen in ALPS patients were profound, suggesting that sirolimus should be considered as a first-line, steroid-sparing treatment of patients needing chronic therapy. The results in other multilineage autoimmune cytopenia cohorts were encouraging, and sirolimus should be considered in children with SLE, ES, and CVID. This trial was registered at www.clinicaltrials.gov as #NCT00392951.
Collapse
Affiliation(s)
- Karen L Bride
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Tiffaney Vincent
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Kim Smith-Whitley
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Michele P Lambert
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Jack J Bleesing
- Division of Bone Marrow Transplantation and Immune Deficiency, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH
| | - Alix E Seif
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Catherine S Manno
- Department of Pediatrics, New York University Langone Medical Center, New York, NY; and
| | - James Casper
- Division of Pediatric Hematology, Oncology and Bone Marrow Transplant, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, WI
| | - Stephan A Grupp
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - David T Teachey
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
41
|
Holcar M, Goropevšek A, Ihan A, Avčin T. Age-Related Differences in Percentages of Regulatory and Effector T Lymphocytes and Their Subsets in Healthy Individuals and Characteristic STAT1/STAT5 Signalling Response in Helper T Lymphocytes. J Immunol Res 2015; 2015:352934. [PMID: 26525134 PMCID: PMC4615205 DOI: 10.1155/2015/352934] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 08/06/2015] [Accepted: 08/27/2015] [Indexed: 01/10/2023] Open
Abstract
The dynamic process of the development of the immune system can in itself result in age-related immune malfunctions. In this study, we analysed lymphocyte subsets in the peripheral blood of 60 healthy donors, divided into groups of children, adolescents, and adults, focusing on effector (Teff) and regulatory (Treg) T lymphocytes and STAT1/STAT5 signalling response in helper T lymphocytes (Th) in adults, using flow cytometry. Our results demonstrate a decrease in the percentage of total Tregs and an increase in the percentage of total Teffs with age and a consequential immense increase in the Teff/Treg ratio. The increase of Teffs was most apparent in Th1, Th1Th17, and Th17CD161- subsets. Significant Th lymphocyte STAT1 expression differences were observed between children and adolescents, which were associated with the decrease in activated Tregs. Higher expression of STAT1 was found in FoxP3hi than in FoxP3low Th lymphocytes, while significant IL-2 induced STAT5 phosphorylation differences were found among the subsets of Th lymphocytes in adults. Our study demonstrates age-related changes in circulating Teff and Treg, as well as significant differences in STAT5/STAT1 signalling among FoxP3+ Th lymphocytes, providing new advances in the understanding of immunosenescence.
Collapse
Affiliation(s)
- Marija Holcar
- Department of Allergology, Rheumatology and Clinical Immunology, University Children's Hospital, University Medical Centre Ljubljana, Bohoričeva 20, SI-1525 Ljubljana, Slovenia
| | - Aleš Goropevšek
- Department of Laboratory Diagnostics, University Medical Centre Maribor, Ljubljanska Ulica 5, SI-2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, SI-2000 Maribor, Slovenia
| | - Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
| | - Tadej Avčin
- Department of Allergology, Rheumatology and Clinical Immunology, University Children's Hospital, University Medical Centre Ljubljana, Bohoričeva 20, SI-1525 Ljubljana, Slovenia
- Department of Pediatrics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
42
|
Autoimmune lymphoproliferative syndrome-like disease in patients with LRBA mutation. Clin Immunol 2015; 159:84-92. [PMID: 25931386 DOI: 10.1016/j.clim.2015.04.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 03/24/2015] [Accepted: 04/19/2015] [Indexed: 12/16/2022]
Abstract
Mutations in LPS-responsive and beige-like anchor (LRBA) gene were recently described in patients with combined immunodeficiency, enteropathy and autoimmune cytopenia. Here, we extend the clinical and immunological phenotypic spectrum of LRBA associated disorders by reporting on three patients from two unrelated families who presented with splenomegaly and lymphadenopathy, cytopenia, elevated double negative T cells and raised serum Fas ligand levels resembling autoimmune lymphoproliferative syndrome (ALPS) and one asymptomatic patient. Homozygous loss of function mutations in LRBA were identified by whole exome analysis. Similar to ALPS patients, Fas mediated apoptosis was impaired in LRBA deficient patients, while apoptosis in response to stimuli of the intrinsic mitochondria mediated apoptotic pathway was even enhanced. This manuscript illustrates the phenotypic overlap of other primary immunodeficiencies with ALPS-like disorders and strongly underlines the necessity of genetic diagnosis in order to provide early correct diagnosis and subsequent care.
Collapse
|
43
|
Eby JM, Kang HK, Klarquist J, Chatterjee S, Mosenson JA, Nishimura MI, Garrett-Mayer E, Longley BJ, Engelhard VH, Mehrotra S, Le Poole IC. Immune responses in a mouse model of vitiligo with spontaneous epidermal de- and repigmentation. Pigment Cell Melanoma Res 2014; 27:1075-85. [PMID: 24935676 PMCID: PMC4470702 DOI: 10.1111/pcmr.12284] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/13/2014] [Indexed: 12/27/2022]
Abstract
To generate a mouse model of spontaneous epidermal depigmentation, parental h3TA2 mice, expressing both a human-derived, tyrosinase-reactive T-cell receptor on T cells and the matching HLA-A2 transgene, were crossed to keratin 14-promoter driven, stem cell factor transgenic (K14-SCF) mice with intra-epidermal melanocytes. In resulting Vitesse mice, spontaneous skin depigmentation precedes symmetrical and sharply demarcated patches of graying hair. Whereas the SCF transgene alone dictates a greater retinoic acid receptor-related orphan receptor gamma (RORγt)(+) T-cell compartment, these cells displayed markedly increased IL-17 expression within Vitesse mice. Similar to patient skin, regulatory T cells were less abundant compared with K14-SCF mice, with the exception of gradually appearing patches of repigmenting skin. The subtle repigmentation observed likely reflects resilient melanocytes that coexist with skin-infiltrating, melanocyte-reactive T cells. Similar repigmenting lesions were found in a different TCR transgenic model of vitiligo developed on an SCF transgenic background, supporting a role for SCF in repigmentation.
Collapse
Affiliation(s)
- Jonathan M Eby
- Oncology Research Institute, Loyola University Chicago, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood 2014; 125:591-9. [PMID: 25359994 DOI: 10.1182/blood-2014-09-602763] [Citation(s) in RCA: 399] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Germline loss-of-function mutations in the transcription factor signal transducer and activator of transcription 3 (STAT3) cause immunodeficiency, whereas somatic gain-of-function mutations in STAT3 are associated with large granular lymphocytic leukemic, myelodysplastic syndrome, and aplastic anemia. Recently, germline mutations in STAT3 have also been associated with autoimmune disease. Here, we report on 13 individuals from 10 families with lymphoproliferation and early-onset solid-organ autoimmunity associated with 9 different germline heterozygous mutations in STAT3. Patients exhibited a variety of clinical features, with most having lymphadenopathy, autoimmune cytopenias, multiorgan autoimmunity (lung, gastrointestinal, hepatic, and/or endocrine dysfunction), infections, and short stature. Functional analyses demonstrate that these mutations confer a gain-of-function in STAT3 leading to secondary defects in STAT5 and STAT1 phosphorylation and the regulatory T-cell compartment. Treatment targeting a cytokine pathway that signals through STAT3 led to clinical improvement in 1 patient, suggesting a potential therapeutic option for such patients. These results suggest that there is a broad range of autoimmunity caused by germline STAT3 gain-of-function mutations, and that hematologic autoimmunity is a major component of this newly described disorder. Some patients for this study were enrolled in a trial registered at www.clinicaltrials.gov as #NCT00001350.
Collapse
|