1
|
Yang W, Xie J, Zhao X, Li X, Liu Q, Sun J, Zhang R, Wei Y, Wang B. Bioinformatics Approach to Investigating the Immuno-Inflammatory Mechanisms of Periodontitis in the Progression of Atherosclerosis. Curr Issues Mol Biol 2025; 47:197. [PMID: 40136451 PMCID: PMC11941604 DOI: 10.3390/cimb47030197] [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: 02/16/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Unstable atherosclerotic plaques are a major cause of acute cardiovascular events and ischemic stroke. Clinical studies have suggested a link between periodontitis and atherosclerotic plaque progression, but the underlying mechanisms remain unclear. To investigate this, transcriptomic datasets related to periodontitis and atherosclerosis were downloaded from Gene Expression Omnibus. A weighted gene co-expression network analysis was used to identify gene modules associated with periodontitis, and the Limma R package identified differentially expressed genes (DEGs) between unstable and stable plaques. Overlapping genes were defined as periodontitis-related DEGs, followed by functional enrichment analysis and protein-protein interaction network construction. Machine learning methods were used to identify biomarkers for unstable plaques related to periodontitis, which were validated using external datasets. Immune infiltration and single-cell analyses were performed to explore the relationship between biomarkers and immune cells. A total of 161 periodontitis-related DEGs were identified, with the pathway analysis showing associations with immune regulation and collagen matrix degradation. HCK, NCKAP1L, and WAS were identified as biomarkers for unstable plaques, demonstrating a high diagnostic value (AUC: 0.9884, 95% CI: 0.9641-1). Immune infiltration analysis revealed an increase in macrophages within unstable plaques. Single-cell analysis showed HCK expression in macrophages and dendritic cells, while NCKAP1L and WAS were expressed in macrophages, dendritic cells, NK cells, and T cells. Consensus clustering identified three expression patterns within unstable plaques. Our findings were validated in atherosclerotic mouse models with periodontitis. This study provides insights into how periodontitis contributes to plaque instability, supporting diagnosis and intervention in patients with periodontitis.
Collapse
Affiliation(s)
- Wenling Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.Y.); (J.X.); (X.L.); (Q.L.); (J.S.); (R.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jianhua Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.Y.); (J.X.); (X.L.); (Q.L.); (J.S.); (R.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xing Zhao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Xuelian Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.Y.); (J.X.); (X.L.); (Q.L.); (J.S.); (R.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qingyi Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.Y.); (J.X.); (X.L.); (Q.L.); (J.S.); (R.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinpeng Sun
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.Y.); (J.X.); (X.L.); (Q.L.); (J.S.); (R.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruiyu Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.Y.); (J.X.); (X.L.); (Q.L.); (J.S.); (R.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.Y.); (J.X.); (X.L.); (Q.L.); (J.S.); (R.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Boyuan Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.Y.); (J.X.); (X.L.); (Q.L.); (J.S.); (R.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
2
|
Vasconcelos-Fontes L, Vieira RC, He M, Ferreira-Reis R, Jurberg AD, Arêas Mendes-da-Cruz D, Andersson J, Cotta-de-Almeida V, Westerberg LS. Controlled WASp activity regulates the proliferative response for Treg cell differentiation in the thymus. Eur J Immunol 2024; 54:e2350450. [PMID: 38356202 DOI: 10.1002/eji.202350450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/16/2024]
Abstract
The Wiskott-Aldrich syndrome protein (WASp) regulates actin cytoskeletal dynamics and function of hematopoietic cells. Mutations in the WAS gene lead to two different syndromes; Wiskott-Aldrich syndrome (WAS) caused by loss-of-function mutations, and X-linked neutropenia (XLN) caused by gain-of-function mutations. We previously showed that WASp-deficient mice have a decreased number of regulatory T (Treg) cells in the thymus and the periphery. We here evaluated the impact of WASp mutations on Treg cells in the thymus of WAS and XLN mouse models. Using in vitro Treg differentiation assays, WAS CD4 single-positive thymocytes have decreased differentiation to Treg cells, despite normal early signaling upon IL-2 and TGF-β stimulation. They failed to proliferate and express CD25 at high levels, leading to poor survival and a lower number of Foxp3+ Treg cells. Conversely, XLN CD4 single-positive thymocytes efficiently differentiate into Foxp3+ Treg cells following a high proliferative response to IL-2 and TGF-β, associated with high CD25 expression when compared with WT cells. Altogether, these results show that specific mutations of WASp affect Treg cell development differently, demonstrating a critical role of WASp activity in supporting Treg cell development and expansion.
Collapse
Affiliation(s)
- Larissa Vasconcelos-Fontes
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Rhaissa C Vieira
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Minghui He
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rafaella Ferreira-Reis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Arnon Dias Jurberg
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Daniella Arêas Mendes-da-Cruz
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - John Andersson
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Karolinska Institutet, Institute of Environmental Medicine, Stockholm, Sweden
| | - Vinicius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Lisa S Westerberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
R P, Shanmugam G, Rakshit S, Sarkar K. Role of Wiskott Aldrich syndrome protein in haematological malignancies: genetics, molecular mechanisms and therapeutic strategies. Pathol Res Pract 2024; 253:155026. [PMID: 38118219 DOI: 10.1016/j.prp.2023.155026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/22/2023]
Abstract
As patients continue to suffer from lymphoproliferative and myeloproliferative diseases known as haematopoietic malignancies can affect the bone marrow, blood, lymph nodes, and lymphatic and non-lymphatic organs. Despite advances in the current treatment, there is still a significant challenge for physicians to improve the therapy of HMs. WASp is an important regulator of actin polymerization and the involvement of WASp in transcription is thought to be linked to the DNA damage response and repair. In some studies, severe immunodeficiency and lymphoid malignancy are caused by WASp mutations or the absence of WASp and these mutations in WAS can alter the function and/or expression of the intracellular protein. Loss-of-function and Gain-of-function mutations in WASp have an impact on cancer malignancies' incidence and onset. Recent studies suggest that depending on the clinical or experimental situation, WASPs and WAVEs can operate as a suppressor or enhancers for cancer malignancy. These dual functions of WASPs and WAVEs in cancer likely arose from their multifaceted role in cells that could be targeted for anticancer drug development. The significant role and their association of WASp in Chronic myeloid leukaemia, Juvenile myelomonocytic leukaemia and T-cell lymphoma is discussed. In this review, we described the structure and function of WASp and its family mechanism, analysing major regulatory effectors and summarising the clinical relevance and drugs that specifically target WASp in disease treatment in various hematopoietic malignancies by different approaches.
Collapse
Affiliation(s)
- Pradeep R
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India
| | - Geetha Shanmugam
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India
| | - Sudeshna Rakshit
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India.
| |
Collapse
|
4
|
Liang F, Peng C, Luo X, Wang L, Huang Y, Yin L, Yue L, Yang J, Zhao X. A single-cell atlas of immunocytes in the spleen of a mouse model of Wiskott-Aldrich syndrome. Cell Immunol 2023; 393-394:104783. [PMID: 37944382 DOI: 10.1016/j.cellimm.2023.104783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/28/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
Wiskott-Aldrich syndrome (WAS) is a disorder characterized by rare X-linked genetic immune deficiency with mutations in the Was gene, which is specifically expressed in hematopoietic cells. The spleen plays a major role in hematopoiesis and red blood cell clearance. However, to date, comprehensive analyses of the spleen in wild-type (WT) and WASp-deficient (WAS-KO) mice, especially at the transcriptome level, have not been reported. In this study, single-cell RNA sequencing (scRNA-seq) was adopted to identify various types of immune cells and investigate the mechanisms underlying immune deficiency. We identified 30 clusters and 10 major cell subtypes among 11,269 cells; these cell types included B cells, T cells, dendritic cells (DCs), natural killer (NK) cells, monocytes, macrophages, granulocytes, stem cells and erythrocytes. Moreover, we evaluated gene expression differences among cell subtypes, identified differentially expressed genes (DEGs), and performed enrichment analyses to identify the reasons for the dysfunction in these different cell populations in WAS. Furthermore, some key genes were identified based on a comparison of the DEGs in each cell type involved in specific and nonspecific immune responses, and further analysis showed that these key genes were previously undiscovered pathology-related genes in WAS-KO mice. In summary, we present a landscape of immune cells in the spleen of WAS-KO mice based on detailed data obtained at single-cell resolution. These unprecedented data revealed the transcriptional characteristics of specific and nonspecific immune cells, and the key genes were identified, laying a foundation for future studies of WAS, especially studies into novel and underexplored mechanisms that may improve gene therapies for WAS.
Collapse
Affiliation(s)
- Fangfang Liang
- Department of Rheumatism and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China; Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Cheng Peng
- Department of Radiology, The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Xianze Luo
- Department of Rheumatism and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Linlin Wang
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Yanyan Huang
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Le Yin
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Luming Yue
- Singleron Biotechnologies, Nanjing, Jiangsu, China
| | - Jun Yang
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China.
| | - Xiaodong Zhao
- Department of Rheumatism and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
5
|
Pille M, Avila J, Sanchez GS, Goetgeluk G, De Munter S, Jansen H, Billiet L, Weening K, Xue H, Bonte S, Ingels J, De Cock L, Pascal E, Deseins L, Kerre T, Taghon T, Leclercq G, Vermijlen D, Davis B, Vandekerckhove B. The Wiskott-Aldrich syndrome protein is required for positive selection during T-cell lineage differentiation. Front Immunol 2023; 14:1188099. [PMID: 37350958 PMCID: PMC10282776 DOI: 10.3389/fimmu.2023.1188099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/15/2023] [Indexed: 06/24/2023] Open
Abstract
The Wiskott-Aldrich syndrome (WAS) is an X-linked primary immune deficiency caused by a mutation in the WAS gene. This leads to altered or absent WAS protein (WASp) expression and function resulting in thrombocytopenia, eczema, recurrent infections, and autoimmunity. In T cells, WASp is required for immune synapse formation. Patients with WAS show reduced numbers of peripheral blood T lymphocytes and an altered T-cell receptor repertoire. In vitro, their peripheral T cells show decreased proliferation and cytokine production upon aCD3/aCD28 stimulation. It is unclear whether these T-cell defects are acquired during peripheral activation or are, in part, generated during thymic development. Here, we assessed the role of WASp during T-cell differentiation using artificial thymic organoid cultures and in the thymus of humanized mice. Although CRISPR/Cas9 WAS knockout hematopoietic stem and progenitor cells (HSPCs) rearranged the T-cell receptor and differentiated to T-cell receptor (TCR)+ CD4+ CD8+ double-positive (DP) cells similar to wild-type HSPCs, a partial defect in the generation of CD8 single-positive (SP) cells was observed, suggesting that WASp is involved in their positive selection. TCR repertoire analysis of the DP and CD8+ SP population, however, showed a polyclonal repertoire with no bias toward autoreactivity. To our knowledge, this is the first study of the role of WASp in human T-cell differentiation and on TCR repertoire generation.
Collapse
Affiliation(s)
- Melissa Pille
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - John Avila
- Brown Foundation Institute of Molecular Medicine, Mc Govern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Glenn Goetgeluk
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Stijn De Munter
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Hanne Jansen
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Lore Billiet
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Karin Weening
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Haipeng Xue
- Brown Foundation Institute of Molecular Medicine, Mc Govern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sarah Bonte
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Joline Ingels
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Laurenz De Cock
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Eva Pascal
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lucas Deseins
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Tessa Kerre
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Tom Taghon
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Georges Leclercq
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Brian Davis
- Brown Foundation Institute of Molecular Medicine, Mc Govern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Bart Vandekerckhove
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
6
|
Wu M, Gao X, Tang Y, Wu W, Zhou J, Shao Y, Hao C, Yang Y, Zhang J. Cbl-b inhibited CD4 + T cell activation by regulating the expression of miR-99a/miR-125b. Int Immunopharmacol 2023; 115:109677. [PMID: 36634415 DOI: 10.1016/j.intimp.2022.109677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023]
Abstract
The molecular regulation of T cell activation has always been a hot topic in immunology. It has been reported that Cbl-b inhibits T cell activation, but the specific molecular mechanism especially for transcriptional regulation has not been very clear so far. Our present study showed that ablation of Cbl-b resulted in the increased expression of miR-99a and miR-125b, and the antagonism of miR-99a or miR-125b could inhibit the Cbl-b-/- T cell over-activation partly. Further study demonstrated that Cbl-b could bind and ubiquitinate SHP-2 in the activated T cells. The activation of SHP-2 deficient T cells was significantly inhibited. Western blot showed that SHP-2 could dephosphorylate HOXA10, and HOXA10 could enter the nucleus under the stimulation of anti-CD3 antibody alone in Cbl-b deficient T cells. Luciferase reporter assay and CUT&Tag qPCR showed that HOXA10 could regulate the expression of miR-99a/miR-125b. Real-time PCR and western blot further indicated that miR-99a/miR-125b functioned on PI3K/AKT pathway to regulate T cell activation. In conclusion, our study demonstrated that Cbl-b ubiquitinated SHP-2 to arrest HOXA10-mediated CD4+ T cell activation by regulating the expression of miR-99a/miR-125b and their function on PI3K/AKT pathway, which might providing a new explanation for the regulation of T cell activation and potential new idea for autoimmune diseases and tumor immunotherapies.
Collapse
Affiliation(s)
- Mengyun Wu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Xiu Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yuxu Tang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Wenyan Wu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Ji Zhou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yu Shao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Chuangli Hao
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, People's Republic of China.
| | - Yi Yang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China.
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China.
| |
Collapse
|
7
|
Liang F, Yang J, Gan Q, Xia Y, Wang L, Huang Y, Peng C. Transcriptomic insights into the role of the spleen in a mouse model of Wiskott‑Aldrich syndrome. Exp Ther Med 2022; 25:64. [PMID: 36605531 PMCID: PMC9798154 DOI: 10.3892/etm.2022.11763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/18/2022] [Indexed: 12/13/2022] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency characterized by microthrombocytopenia, eczema, recurrent infection and increased incidence of autoimmune disorders and malignancy. WAS is caused by mutations in the was gene, which is expressed exclusively in hematopoietic cells; the spleen serves an important role in hematopoiesis and red blood cell clearance. However, to the best of our knowledge, detailed comparative analysis of the spleen between WASp-knockout (WAS-KO) and wild-type (WT) mice, particularly at the transcriptomic level, have not been reported. The present study investigated the differences in the transcriptomes of spleen tissue of 10-week-old WAS-KO mice. Comparison of the gene expression profiles of WAS-KO and WT mice revealed 1,964 differentially expressed genes (DEGs). Among these genes, 996 DEGs were upregulated and 968 were downregulated in WAS-KO mice. To determine the functions of DEGs, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed for significantly upregulated and downregulated DEGs. The results showed that the levels of cell senescence and apoptosis-associated genes were increased, antigen processing and presentation mechanisms involved in the immune response were damaged and signal transduction processes were impaired in the spleen of WAS-KO mice. Thus, was gene deletion may lead to anemia and hemolysis-associated disease, primarily due to increased osmotic fragility of red blood cells, low hemoglobin and increased bilirubin levels and serum ferritin. These results indicated that senescence and apoptosis of blood cells also play an important role in the occurrence of WAS. Therefore, the present findings provide a theoretical basis for further study to improve the treatment of WAS.
Collapse
Affiliation(s)
- Fangfang Liang
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, Guangdong 518034, P.R. China
| | - Jun Yang
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, Guangdong 518034, P.R. China
| | - Qian Gan
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, Guangdong 518034, P.R. China
| | - Yu Xia
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, Guangdong 518034, P.R. China
| | - Linlin Wang
- Department of Radiology, The Third People's Hospital of Shenzhen, Shenzhen, Guangdong 518112, P.R. China
| | - Yanyan Huang
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, Guangdong 518034, P.R. China
| | - Cheng Peng
- Department of Radiology, The Third People's Hospital of Shenzhen, Shenzhen, Guangdong 518112, P.R. China,Correspondence to: Dr Cheng Peng, Department of Radiology, The Third People's Hospital of Shenzhen, 29 Bulan Road, Shenzhen, Guangdong 518112, P.R. China
| |
Collapse
|
8
|
Novel insights into mouse models of ectopic proplatelet release. Blood Adv 2022; 6:6135-6139. [PMID: 36251748 PMCID: PMC9768245 DOI: 10.1182/bloodadvances.2022007824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/01/2022] [Accepted: 09/04/2022] [Indexed: 12/31/2022] Open
Abstract
Mature bone marrow (BM) megakaryocytes (MKs) produce platelets by extending proplatelets into sinusoidal blood vessels. Defects in this process can lead to thrombocytopenia and increased risk of bleeding. Mice lacking the actin-regulatory proteins Profilin 1 (PFN1), Wiskott-Aldrich Syndrome protein (WASp), Actin Related Protein 2/3 complex (Arp2/3), or adhesion and degranulation-promoting adapter protein (ADAP) display thrombocytopenia and ectopic release of (pro)platelet-like particles into the BM compartment, pointing to an important axis of actin-mediated directional proplatelet formation. The mechanism underlying ectopic release in these mice is still not completely understood. However, we hypothesized that similar functional defects account for this observation. We analyzed WASp-, ADAP-, PFN1-, and ARPC2-knockout mice to determine the role of actin reorganization and integrin activation in directional proplatelet formation. ADAP-, ARPC2-, and PFN1-deficient MKs displayed reduced adhesion to collagen, defective F-actin organization, and diminished β1-integrin activation. WASp-deficient MKs showed the strongest reduction in the adhesion assay of collagen and altered F-actin organization with reduced podosome formation. Our results indicate that ADAP, PFN1, WASp, and ARP2/3 are part of the same pathway that regulates polarization processes in MKs and directional proplatelet formation into BM sinusoids.
Collapse
|
9
|
Sharma TT, Rabizadeh RR, Prabhakar VS, Bury MI, Sharma AK. Evolving Experimental Platforms to Evaluate Ulcerative Colitis. Adv Biol (Weinh) 2022; 6:e2200018. [PMID: 35866469 DOI: 10.1002/adbi.202200018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/06/2022] [Indexed: 01/28/2023]
Abstract
Ulcerative colitis (UC) is a multifactorial disease defined by chronic intestinal inflammation with idiopathic origins. It has a predilection to affect the mucosal lining of the large intestines and rectum. Management of UC depends upon numerous factors that include disease pathogenesis and severity that are maintained via medical or surgical means. Chronic inflammation that is left untreated or managed poorly from a clinical stance can result in intestinal ulceration accompanied by resulting physiological dysfunction. End-stage UC is mediated by surgical intervention with the resection of diseased tissue. This can lead to numerous health-related quality of life issues but is considered a curative approach. Regimens to treat UC are ever evolving and find their basis within various platforms to evaluate and treat UC. Numerous modeling systems have been examined to delineate potential mechanisms of action. However, UC is a heterogenous disease spanning unknown genetic origins coupled with environmental factors that can influence disease outcomes and related treatment procedures. Unfortunately, there is no one-size-fits-all model to fully assess all facets of UC. Within the context of this review article, the utility of various approaches that have been employed to gain insight into different aspects of UC will be investigated.
Collapse
Affiliation(s)
- Tiffany T Sharma
- Lurie Children's Hospital, Division of Pediatric Urology, Chicago, IL, 60611, USA.,Stanley Manne Children's Research Institute, Chicago, IL, 60611, USA
| | - Rebecca R Rabizadeh
- Lurie Children's Hospital, Division of Pediatric Urology, Chicago, IL, 60611, USA
| | - Vibhav S Prabhakar
- Lurie Children's Hospital, Division of Pediatric Urology, Chicago, IL, 60611, USA
| | - Matthew I Bury
- Lurie Children's Hospital, Division of Pediatric Urology, Chicago, IL, 60611, USA
| | - Arun K Sharma
- Lurie Children's Hospital, Division of Pediatric Urology, Chicago, IL, 60611, USA.,Stanley Manne Children's Research Institute, Chicago, IL, 60611, USA.,Feinberg School of Medicine, Department of Urology, Northwestern University, Chicago, IL, 60611, USA.,McCormick School of Engineering, Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA.,Center for Advanced Regenerative Engineering (CARE), Northwestern University, Evanston, IL, 60208, USA.,Simpson Querrey Institute (SQI), Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
10
|
Yuan B, Zhou X, Suzuki K, Ramos-Mandujano G, Wang M, Tehseen M, Cortés-Medina LV, Moresco JJ, Dunn S, Hernandez-Benitez R, Hishida T, Kim NY, Andijani MM, Bi C, Ku M, Takahashi Y, Xu J, Qiu J, Huang L, Benner C, Aizawa E, Qu J, Liu GH, Li Z, Yi F, Ghosheh Y, Shao C, Shokhirev M, Comoli P, Frassoni F, Yates JR, Fu XD, Esteban CR, Hamdan S, Li M, Izpisua Belmonte JC. Wiskott-Aldrich syndrome protein forms nuclear condensates and regulates alternative splicing. Nat Commun 2022; 13:3646. [PMID: 35752626 PMCID: PMC9233711 DOI: 10.1038/s41467-022-31220-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/06/2022] [Indexed: 11/09/2022] Open
Abstract
The diverse functions of WASP, the deficiency of which causes Wiskott-Aldrich syndrome (WAS), remain poorly defined. We generated three isogenic WAS models using patient induced pluripotent stem cells and genome editing. These models recapitulated WAS phenotypes and revealed that WASP deficiency causes an upregulation of numerous RNA splicing factors and widespread altered splicing. Loss of WASP binding to splicing factor gene promoters frequently leads to aberrant epigenetic activation. WASP interacts with dozens of nuclear speckle constituents and constrains SRSF2 mobility. Using an optogenetic system, we showed that WASP forms phase-separated condensates that encompasses SRSF2, nascent RNA and active Pol II. The role of WASP in gene body condensates is corroborated by ChIPseq and RIPseq. Together our data reveal that WASP is a nexus regulator of RNA splicing that controls the transcription of splicing factors epigenetically and the dynamics of the splicing machinery through liquid-liquid phase separation.
Collapse
Affiliation(s)
- Baolei Yuan
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Xuan Zhou
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Keiichiro Suzuki
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Institute for Advanced Co-Creation Studies, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Gerardo Ramos-Mandujano
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Mengge Wang
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Muhammad Tehseen
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Lorena V Cortés-Medina
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - James J Moresco
- Department of Cell Biology, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Sarah Dunn
- The Waitt Advanced Biophotonics Core Facility, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Reyna Hernandez-Benitez
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Altos Labs, Inc. 5510 Morehouse Drive, Suite 300, San Diego, CA, 92121, USA
| | - Tomoaki Hishida
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Laboratory of Biological Chemistry, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shitibancho, Wakayama, Wakayama, 640-8156, Japan
| | - Na Young Kim
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Manal M Andijani
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Chongwei Bi
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Manching Ku
- Next-generation sequencing core, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Yuta Takahashi
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Jinna Xu
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Jinsong Qiu
- Department of Cellular & Molecular Medicine, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Ling Huang
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Christopher Benner
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Emi Aizawa
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Institute for Advanced Co-Creation Studies, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Jing Qu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhongwei Li
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,University of Southern California, 1333 San Pablo Street, MMR 618, Los Angeles, CA, 90033, USA
| | - Fei Yi
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Ambys Medicines, 131 Oyster Point Blvd. Suite 200, South San Francisco, CA, 94080, USA
| | - Yanal Ghosheh
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Changwei Shao
- Department of Cellular & Molecular Medicine, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Maxim Shokhirev
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Patrizia Comoli
- Pediatric Hematology/Oncology and Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Francesco Frassoni
- Department of Research Laboratories and Director of Center for Stem Cell and Cell Therapy, Instituto G. Gaslini Children Hospital Scientific Institute, 16147, Genova, Italy
| | - John R Yates
- Department of Cell Biology, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Xiang-Dong Fu
- Department of Cellular & Molecular Medicine, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Concepcion Rodriguez Esteban
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Altos Labs, Inc. 5510 Morehouse Drive, Suite 300, San Diego, CA, 92121, USA
| | - Samir Hamdan
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Mo Li
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
| | - Juan Carlos Izpisua Belmonte
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia. .,Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA. .,Altos Labs, Inc. 5510 Morehouse Drive, Suite 300, San Diego, CA, 92121, USA.
| |
Collapse
|
11
|
Descatoire M, Fritzen R, Rotman S, Kuntzelman G, Leber XC, Droz-Georget S, Thrasher AJ, Traggiai E, Candotti F. Critical role of WASp in germinal center tolerance through regulation of B cell apoptosis and diversification. Cell Rep 2022; 38:110474. [PMID: 35263577 DOI: 10.1016/j.celrep.2022.110474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/18/2021] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Abstract
A main feature of Wiskott-Aldrich syndrome (WAS) is increased susceptibility to autoimmunity. A key contribution of B cells to development of these complications has been demonstrated through studies of samples from affected individuals and mouse models of the disease, but the role of the WAS protein (WASp) in controlling peripheral tolerance has not been specifically explored. Here we show that B cell responses remain T cell dependent in constitutive WASp-deficient mice, whereas selective WASp deletion in germinal center B cells (GCBs) is sufficient to induce broad development of self-reactive antibodies and kidney pathology, pointing to loss of germinal center tolerance as a primary cause leading to autoimmunity. Mechanistically, we show that WASp is upregulated in GCBs and regulates apoptosis and plasma cell differentiation in the germinal center and that the somatic hypermutation-derived diversification is the basis of autoantibody development.
Collapse
Affiliation(s)
- Marc Descatoire
- Laboratory of Inherited Immune Disorders, Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | | | - Samuel Rotman
- Service of Clinical Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | | | - Stephanie Droz-Georget
- Laboratory of Inherited Immune Disorders, Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Adrian J Thrasher
- University College of London, Great Ormond Street Institute of Child Health, London, UK
| | | | - Fabio Candotti
- Laboratory of Inherited Immune Disorders, Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
12
|
Khanna C, Le Coz C, Vaccaro C, Pillarisetti P, Knox AVC, Sy A, Behrens EM, Buchbinder D, Romberg N. Lymphocytes Utilize Somatic Mutations, Epigenetic Silencing, and the Proteasome to Escape Truncated WASP Expression. J Clin Immunol 2022; 42:753-759. [PMID: 35149963 DOI: 10.1007/s10875-022-01224-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/28/2022] [Indexed: 11/25/2022]
Abstract
Wiskott-Aldrich Syndrome Protein (WASP) deficiency causes Wiskott-Aldrich Syndrome (WAS), a sex-linked disorder characterized by combined immunodeficiency, microthrombocytopenia, and eczema. Like WASP-deficient humans, WASP-deficient mice produce normal numbers of functionally defective T cells. Here, we report a WAS patient with a novel germline frameshifting WAS mutation encoding a truncated form of WASP lacking the C-terminal cofilin homology (C) and the acidic region (A) domains (WASPΔCA). Although stably overexpressed in embryonic kidney cell lines, WASPΔCA was undetectable in circulating patient leukocytes. Deep sequencing, transcript profiling, and protein degradation analyses demonstrated patient lymphocytes employ an array of genetic, epigenetic, and proteasomal strategies to avoid expressing WASPΔCA.
Collapse
Affiliation(s)
- Caroline Khanna
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Carole Le Coz
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Courtney Vaccaro
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Piyush Pillarisetti
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ainsley V C Knox
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew Sy
- Department of Hematology, The Children's Hospital of Orange County, Orange, CA, USA
| | - Edward M Behrens
- Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Buchbinder
- Department of Hematology, The Children's Hospital of Orange County, Orange, CA, USA
| | - Neil Romberg
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Leonard and Madlyn Abramson Pediatric Research Center, 3615 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
13
|
Shah K, Al-Haidari A, Sun J, Kazi JU. T cell receptor (TCR) signaling in health and disease. Signal Transduct Target Ther 2021; 6:412. [PMID: 34897277 PMCID: PMC8666445 DOI: 10.1038/s41392-021-00823-w] [Citation(s) in RCA: 227] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Interaction of the T cell receptor (TCR) with an MHC-antigenic peptide complex results in changes at the molecular and cellular levels in T cells. The outside environmental cues are translated into various signal transduction pathways within the cell, which mediate the activation of various genes with the help of specific transcription factors. These signaling networks propagate with the help of various effector enzymes, such as kinases, phosphatases, and phospholipases. Integration of these disparate signal transduction pathways is done with the help of adaptor proteins that are non-enzymatic in function and that serve as a scaffold for various protein-protein interactions. This process aids in connecting the proximal to distal signaling pathways, thereby contributing to the full activation of T cells. This review provides a comprehensive snapshot of the various molecules involved in regulating T cell receptor signaling, covering both enzymes and adaptors, and will discuss their role in human disease.
Collapse
Affiliation(s)
- Kinjal Shah
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Amr Al-Haidari
- Clinical Genetics and Pathology, Skåne University Hospital, Region Skåne, Lund, Sweden
- Clinical Sciences Department, Surgery Research Unit, Lund University, Malmö, Sweden
| | - Jianmin Sun
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Julhash U Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
14
|
Tsou AM, Goettel JA, Bao B, Biswas A, Kang YH, Redhu NS, Peng K, Putzel GG, Saltzman J, Kelly R, Gringauz J, Barends J, Hatazaki M, Frei SM, Emani R, Huang Y, Shen Z, Fox JG, Glickman JN, Horwitz BH, Snapper SB. Utilizing a reductionist model to study host-microbe interactions in intestinal inflammation. MICROBIOME 2021; 9:215. [PMID: 34732258 PMCID: PMC8565002 DOI: 10.1186/s40168-021-01161-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/10/2021] [Indexed: 05/10/2023]
Abstract
BACKGROUND The gut microbiome is altered in patients with inflammatory bowel disease, yet how these alterations contribute to intestinal inflammation is poorly understood. Murine models have demonstrated the importance of the microbiome in colitis since colitis fails to develop in many genetically susceptible animal models when re-derived into germ-free environments. We have previously shown that Wiskott-Aldrich syndrome protein (WASP)-deficient mice (Was-/-) develop spontaneous colitis, similar to human patients with loss-of-function mutations in WAS. Furthermore, we showed that the development of colitis in Was-/- mice is Helicobacter dependent. Here, we utilized a reductionist model coupled with multi-omics approaches to study the role of host-microbe interactions in intestinal inflammation. RESULTS Was-/- mice colonized with both altered Schaedler flora (ASF) and Helicobacter developed colitis, while those colonized with either ASF or Helicobacter alone did not. In Was-/- mice, Helicobacter relative abundance was positively correlated with fecal lipocalin-2 (LCN2), a marker of intestinal inflammation. In contrast, WT mice colonized with ASF and Helicobacter were free of inflammation and strikingly, Helicobacter relative abundance was negatively correlated with LCN2. In Was-/- colons, bacteria breach the mucus layer, and the mucosal relative abundance of ASF457 Mucispirillum schaedleri was positively correlated with fecal LCN2. Meta-transcriptomic analyses revealed that ASF457 had higher expression of genes predicted to enhance fitness and immunogenicity in Was-/- compared to WT mice. In contrast, ASF519 Parabacteroides goldsteinii's relative abundance was negatively correlated with LCN2 in Was-/- mice, and transcriptional analyses showed lower expression of genes predicted to facilitate stress adaptation by ASF519 in Was-/-compared to WT mice. CONCLUSIONS These studies indicate that the effect of a microbe on the immune system can be context dependent, with the same bacteria eliciting a tolerogenic response under homeostatic conditions but promoting inflammation in immune-dysregulated hosts. Furthermore, in inflamed environments, some bacteria up-regulate genes that enhance their fitness and immunogenicity, while other bacteria are less able to adapt and decrease in abundance. These findings highlight the importance of studying host-microbe interactions in different contexts and considering how the transcriptional profile and fitness of bacteria may change in different hosts when developing microbiota-based therapeutics. Video abstract.
Collapse
Affiliation(s)
- Amy M Tsou
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, New York, NY, USA.
- Division of Pediatric Gastroenterology and Nutrition, Weill Cornell Medical College, New York, NY, USA.
| | - Jeremy A Goettel
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bin Bao
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Amlan Biswas
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yu Hui Kang
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Naresh S Redhu
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kaiyue Peng
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai, China
| | - Gregory G Putzel
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, New York, NY, USA
| | - Jeffrey Saltzman
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Ryan Kelly
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Jordan Gringauz
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Jared Barends
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Mai Hatazaki
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, New York, NY, USA
| | - Sandra M Frei
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rohini Emani
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ying Huang
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai, China
| | - Zeli Shen
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jonathan N Glickman
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Bruce H Horwitz
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
15
|
Sudhakar M, Rikhi R, Loganathan SK, Suri D, Singh S. Autoimmunity in Wiskott-Aldrich Syndrome: Updated Perspectives. APPLICATION OF CLINICAL GENETICS 2021; 14:363-388. [PMID: 34447261 PMCID: PMC8384432 DOI: 10.2147/tacg.s213920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/18/2021] [Indexed: 11/23/2022]
Abstract
Wiskott–Aldrich syndrome (WAS) is an uncommon X-linked combined-immunodeficiency disorder characterized by a triad of thrombocytopenia, eczema, and immunodeficiency. Patients with WAS are also predisposed to autoimmunity and malignancy. Autoimmune manifestations have been reported in 26%–72% of patients with WAS. Autoimmunity is an independent predictor of poor prognosis and predisposes to malignancy. Development of autoimmunity is also an early pointer of the need for hematopoietic stem–cell transplantation. In this manuscript, we have collated the published data and present a narrative review on autoimmune manifestations in WAS. A summary of currently proposed immunopathogenic mechanisms and genetic variants associated with development of autoimmunity in WAS is also included.
Collapse
Affiliation(s)
- Murugan Sudhakar
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rashmi Rikhi
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sathish Kumar Loganathan
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepti Suri
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Surjit Singh
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
16
|
Oliveira MMS, Kung SY, Moreau HD, Maurin M, Record J, Sanséau D, Nylén S, Lennon-Duménil AM, Westerberg LS. The WASp L272P gain-of-function mutation alters dendritic cell coordination of actin dynamics for migration and adhesion. J Leukoc Biol 2021; 111:793-803. [PMID: 34431547 DOI: 10.1002/jlb.1ab0821-013rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) devoid of the actin regulator Wiskott-Aldrich syndrome protein (WASp) show reduced directed migration and decreased formation of podosome adhesion structures. We examined DCs expressing a gain-of-function mutation in WASp, WASp L272P, identified in X-linked neutropenia patients. Analysis of WASp L272P DCs was compared to WASp-deficient DCs to examine how WASp activity influences DC migratory responses. In confined space, WASp-deficient DCs had increased migration speed whereas WASp L272P DCs had similar average speed but increased speed fluctuations, reduced displacement, and atypical rounded morphology, compared to wild-type (WT) DCs. Using an ear inflammation model and flow cytometry analysis, WT, WASp-deficient, and WASp L272P DCs were found to migrate in comparable numbers to the draining lymph nodes (LNs). However, histology analysis revealed that migratory DCs of WASp deficient and WASp L272P mice were mainly located in the collagenous capsule of the LN whereas WT DCs were located inside the LN. Analysis of ultrastructural features revealed that WASp L272P DCs had reduced cell area but formed larger podosome structures when compared to WT DCs. Together, our data suggest that WASp activity regulates DC migration and that loss-of-function and gain-of-function in WASp activity lead to different and phenotype-specific DC migratory behavior.
Collapse
Affiliation(s)
- Mariana M S Oliveira
- Department of Microbiology Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Shin-Yu Kung
- Department of Microbiology Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | | | - Mathieu Maurin
- Institute Curie, PSL Research University, INSERM U932, Paris, France
| | - Julien Record
- Department of Microbiology Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | | | - Susanne Nylén
- Department of Microbiology Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | | | - Lisa S Westerberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
17
|
Jacobs HM, Arkatkar T, Du SW, Scharping NE, Woods J, Li QZ, Hudkins KL, Alpers CE, Rawlings DJ, Jackson SW. TACI haploinsufficiency protects against BAFF-driven humoral autoimmunity in mice. Eur J Immunol 2021; 51:2225-2236. [PMID: 34146342 DOI: 10.1002/eji.202149244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/26/2021] [Accepted: 06/16/2021] [Indexed: 11/06/2022]
Abstract
Polymorphisms in TACI, a BAFF family cytokine receptor, are linked to diverse human immune disorders including common variable immunodeficiency (CVID) and systemic lupus erythematosus (SLE). Functional studies of individual variants show modest impacts on surface TACI expression and/or downstream signal transduction, indicating that relatively subtle variation in TACI activity can impact human B-cell biology. However, significant complexity underlies TACI biology, including both positive and negative regulation of physiologic and pathogenic B-cell responses. To model these contradictory events, we compared the functional impact of TACI deletion on separate models of murine SLE driven by T cell-independent and -dependent breaks in B-cell tolerance. First, we studied whether reduced surface TACI expression was sufficient to protect against progressive BAFF-mediated systemic autoimmunity. Strikingly, despite a relatively modest impact on surface TACI levels, TACI haploinsufficiency markedly reduced pathogenic RNA-associated autoantibody titers and conferred long-term protection from BAFF-driven lupus nephritis. In contrast, B cell-intrinsic TACI deletion exerted a limited impact of autoantibody generation in murine lupus characterized by spontaneous germinal center formation and T cell-dependent humoral autoimmunity. Together, these combined data provide new insights into TACI biology and highlight how TACI signals must be tightly regulated during protective and pathogenic B-cell responses.
Collapse
Affiliation(s)
| | | | - Samuel W Du
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | - Quan-Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kelly L Hudkins
- Departments of Laboratory Medicine and Pathology, Immunology and Pediatrics, University of Washington School of Medicine, Washington, WA, USA
| | - Charles E Alpers
- Departments of Laboratory Medicine and Pathology, Immunology and Pediatrics, University of Washington School of Medicine, Washington, WA, USA
| | | | | |
Collapse
|
18
|
Heib T, Hermanns HM, Manukjan G, Englert M, Kusch C, Becker IC, Gerber A, Wackerbarth LM, Burkard P, Dandekar T, Balkenhol J, Jahn D, Beck S, Meub M, Dütting S, Stigloher C, Sauer M, Cherpokova D, Schulze H, Brakebusch C, Nieswandt B, Nagy Z, Pleines I. RhoA/Cdc42 signaling drives cytoplasmic maturation but not endomitosis in megakaryocytes. Cell Rep 2021; 35:109102. [PMID: 33979620 DOI: 10.1016/j.celrep.2021.109102] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/20/2021] [Accepted: 04/18/2021] [Indexed: 12/15/2022] Open
Abstract
Megakaryocytes (MKs), the precursors of blood platelets, are large, polyploid cells residing mainly in the bone marrow. We have previously shown that balanced signaling of the Rho GTPases RhoA and Cdc42 is critical for correct MK localization at bone marrow sinusoids in vivo. Using conditional RhoA/Cdc42 double-knockout (DKO) mice, we reveal here that RhoA/Cdc42 signaling is dispensable for the process of polyploidization in MKs but essential for cytoplasmic MK maturation. Proplatelet formation is virtually abrogated in the absence of RhoA/Cdc42 and leads to severe macrothrombocytopenia in DKO animals. The MK maturation defect is associated with downregulation of myosin light chain 2 (MLC2) and β1-tubulin, as well as an upregulation of LIM kinase 1 and cofilin-1 at both the mRNA and protein level and can be linked to impaired MKL1/SRF signaling. Our findings demonstrate that MK endomitosis and cytoplasmic maturation are separately regulated processes, and the latter is critically controlled by RhoA/Cdc42.
Collapse
Affiliation(s)
- Tobias Heib
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Heike M Hermanns
- Department of Internal Medicine II, Hepatology Research Laboratory, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Georgi Manukjan
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Maximilian Englert
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Charly Kusch
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Isabelle Carlotta Becker
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Annika Gerber
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Lou Martha Wackerbarth
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Philipp Burkard
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Johannes Balkenhol
- Department of Internal Medicine II, Hepatology Research Laboratory, University Hospital Würzburg, 97080 Würzburg, Germany; Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Daniel Jahn
- Department of Internal Medicine II, Hepatology Research Laboratory, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Sarah Beck
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Mara Meub
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Sebastian Dütting
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Christian Stigloher
- Imaging Core Facility, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Deya Cherpokova
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Harald Schulze
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Cord Brakebusch
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany.
| | - Zoltan Nagy
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Irina Pleines
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany.
| |
Collapse
|
19
|
Lymphopenia, Lymphopenia-Induced Proliferation, and Autoimmunity. Int J Mol Sci 2021; 22:ijms22084152. [PMID: 33923792 PMCID: PMC8073364 DOI: 10.3390/ijms22084152] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
Immune homeostasis is a tightly regulated system that is critical for defense against invasion by foreign pathogens and protection from self-reactivity for the survival of an individual. How the defects in this system might result in autoimmunity is discussed in this review. Reduced lymphocyte number, termed lymphopenia, can mediate lymphopenia-induced proliferation (LIP) to maintain peripheral lymphocyte numbers. LIP not only occurs in normal physiological conditions but also correlates with autoimmunity. Of note, lymphopenia is also a typical marker of immune aging, consistent with the fact that not only the autoimmunity increases in the elderly, but also autoimmune diseases (ADs) show characteristics of immune aging. Here, we discuss the types and rates of LIP in normal and autoimmune conditions, as well as the coronavirus disease 2019 in the context of LIP. Importantly, although the causative role of LIP has been demonstrated in the development of type 1 diabetes and rheumatoid arthritis, a two-hit model has suggested that the factors other than lymphopenia are required to mediate the loss of control over homeostasis to result in ADs. Interestingly, these factors may be, if not totally, related to the function/number of regulatory T cells which are key modulators to protect from self-reactivity. In this review, we summarize the important roles of lymphopenia/LIP and the Treg cells in various autoimmune conditions, thereby highlighting them as key therapeutic targets for autoimmunity treatments.
Collapse
|
20
|
Fragliasso V, Tameni A, Inghirami G, Mularoni V, Ciarrocchi A. Cytoskeleton Dynamics in Peripheral T Cell Lymphomas: An Intricate Network Sustaining Lymphomagenesis. Front Oncol 2021; 11:643620. [PMID: 33928032 PMCID: PMC8076600 DOI: 10.3389/fonc.2021.643620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/17/2021] [Indexed: 12/04/2022] Open
Abstract
Defects in cytoskeleton functions support tumorigenesis fostering an aberrant proliferation and promoting inappropriate migratory and invasive features. The link between cytoskeleton and tumor features has been extensively investigated in solid tumors. However, the emerging genetic and molecular landscape of peripheral T cell lymphomas (PTCL) has unveiled several alterations targeting structure and function of the cytoskeleton, highlighting its role in cell shape changes and the aberrant cell division of malignant T cells. In this review, we summarize the most recent evidence about the role of cytoskeleton in PTCLs development and progression. We also discuss how aberrant signaling pathways, like JAK/STAT3, NPM-ALK, RhoGTPase, and Aurora Kinase, can contribute to lymphomagenesis by modifying the structure and the signaling properties of cytoskeleton.
Collapse
Affiliation(s)
- Valentina Fragliasso
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Annalisa Tameni
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Valentina Mularoni
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
21
|
Kritikou JS, Oliveira MM, Record J, Saeed MB, Nigam SM, He M, Keszei M, Wagner AK, Brauner H, Sendel A, Sedimbi SK, Rentouli S, Lane DP, Snapper SB, Kärre K, Vandenberghe P, Orange JS, Westerberg LS. Constitutive activation of WASp leads to abnormal cytotoxic cells with increased granzyme B and degranulation response to target cells. JCI Insight 2021; 6:140273. [PMID: 33621210 PMCID: PMC8026198 DOI: 10.1172/jci.insight.140273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 02/17/2021] [Indexed: 11/23/2022] Open
Abstract
X-linked neutropenia (XLN) is caused by gain-of-function mutations in the actin regulator Wiskott-Aldrich Syndrome protein (WASp). XLN patients have reduced numbers of cytotoxic cells in peripheral blood; however, their capacity to kill tumor cells remains to be determined. Here, we examined NK and T cells from 2 patients with XLN harboring the activating WASpL270P mutation. XLN patient NK and T cells had increased granzyme B content and elevated degranulation and IFN-γ production when compared with healthy control cells. Murine WASpL272P NK and T cells formed stable synapses with YAC-1 tumor cells and anti-CD3/CD28-coated beads, respectively. WASpL272P mouse T cells had normal degranulation and cytokine response whereas WASpL272P NK cells showed an enhanced response. Imaging experiments revealed that while WASpL272P CD8+ T cells had increased accumulation of actin upon TCR activation, WASpL272P NK cells had normal actin accumulation at lytic synapses triggered through NKp46 signaling but had impaired response to lymphocyte function associated antigen-1 engagement. When compared with WT mice, WASpL272P mice showed reduced growth of B16 melanoma and increased capacity to reject MHC class I-deficient cells. Together, our data suggest that cytotoxic cells with constitutively active WASp have an increased capacity to respond to and kill tumor cells.
Collapse
Affiliation(s)
| | | | - Julien Record
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
| | - Mezida B. Saeed
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
| | - Saket M. Nigam
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
| | - Minghui He
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
| | - Marton Keszei
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
| | - Arnika K. Wagner
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
| | - Hanna Brauner
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
- Department of Medicine, Solna, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anton Sendel
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
| | | | | | - David P. Lane
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
| | - Scott B. Snapper
- Gastroenterology Division, Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Klas Kärre
- Department of Microbiology Tumor and Cell Biology, Biomedicum C7, and
| | | | - Jordan S. Orange
- Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | | |
Collapse
|
22
|
Wang C, Sample KM, Gajendran B, Kapranov P, Liu W, Hu A, Zacksenhaus E, Li Y, Hao X, Ben-David Y. FLI1 Induces Megakaryopoiesis Gene Expression Through WAS/WIP-Dependent and Independent Mechanisms; Implications for Wiskott-Aldrich Syndrome. Front Immunol 2021; 12:607836. [PMID: 33717090 PMCID: PMC7953068 DOI: 10.3389/fimmu.2021.607836] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Wiskott–Aldrich Syndrome, WAS/WAVE, is a rare, X-linked immune-deficiency disease caused by mutations in the WAS gene, which together with its homolog, N-WASP, regulates actin cytoskeleton remodeling and cell motility. WAS patients suffer from microthrombocytopenia, characterized by a diminished number and size of platelets, though the underlying mechanism is not fully understood. Here, we identified FLI1 as a direct transcriptional regulator of WAS and its binding partner WIP. Depletion of either WAS or WIP in human erythroleukemic cells accelerated cell proliferation, suggesting tumor suppressor function of both genes in leukemia. Depletion of WAS/WIP also led to a significant reduction in the percentage of CD41 and CD61 positive cells, which mark committed megakaryocytes. RNAseq analysis revealed common changes in megakaryocytic gene expression following FLI1 or WASP knockdown. However, in contrast to FLI1, WASP depletion did not alter expression of late-stage platelet-inducing genes. N-WASP was not regulated by FLI1, yet its silencing also reduced the percentage of CD41+ and CD61+ megakaryocytes. Moreover, combined knockdown of WASP and N-WASP further suppressed megakaryocyte differentiation, indicating a major cooperation of these related genes in controlling megakaryocytic cell fate. However, unlike WASP/WIP, N-WASP loss suppressed leukemic cell proliferation. WASP, WIP and N-WASP depletion led to induction of FLI1 expression, mediated by GATA1, and this may mitigate the severity of platelet deficiency in WAS patients. Together, these results uncover a crucial role for FLI1 in megakaryocyte differentiation, implicating this transcription factor in regulating microthrombocytopenia associated with Wiskott–Aldrich syndrome.
Collapse
Affiliation(s)
- Chunlin Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Klarke M Sample
- The National Health Commission's Key Laboratory of Immunological Pulmonary Disease, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou University, Guiyang, China
| | - Babu Gajendran
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Philipp Kapranov
- School of Biomedical Sciences, Institute of Genomics, Huaqiao University, Xiamen, China
| | - Wuling Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Anling Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Eldad Zacksenhaus
- Department of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Advanced Diagnostics, Toronto General Research Institute-University Health Network, Toronto, ON, Canada
| | - Yanmei Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Xiaojiang Hao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| |
Collapse
|
23
|
Vainchenker W, Arkoun B, Basso-Valentina F, Lordier L, Debili N, Raslova H. Role of Rho-GTPases in megakaryopoiesis. Small GTPases 2021; 12:399-415. [PMID: 33570449 PMCID: PMC8583283 DOI: 10.1080/21541248.2021.1885134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Megakaryocytes (MKs) are the bone marrow (BM) cells that generate blood platelets by a process that requires: i) polyploidization responsible for the increased MK size and ii) cytoplasmic organization leading to extension of long pseudopods, called proplatelets, through the endothelial barrier to allow platelet release into blood. Low level of localized RHOA activation prevents actomyosin accumulation at the cleavage furrow and participates in MK polyploidization. In the platelet production, RHOA and CDC42 play opposite, but complementary roles. RHOA inhibits both proplatelet formation and MK exit from BM, whereas CDC42 drives the development of the demarcation membranes and MK migration in BM. Moreover, the RhoA or Cdc42 MK specific knock-out in mice and the genetic alterations in their down-stream effectors in human induce a thrombocytopenia demonstrating their key roles in platelet production. A better knowledge of Rho-GTPase signalling is thus necessary to develop therapies for diseases associated with platelet production defects. Abbreviations: AKT: Protein Kinase BARHGEF2: Rho/Rac Guanine Nucleotide Exchange Factor 2ARP2/3: Actin related protein 2/3BM: Bone marrowCDC42: Cell division control protein 42 homologCFU-MK: Colony-forming-unit megakaryocyteCIP4: Cdc42-interacting protein 4mDIA: DiaphanousDIAPH1; Protein diaphanous homolog 1ECT2: Epithelial Cell Transforming Sequence 2FLNA: Filamin AGAP: GTPase-activating proteins or GTPase-accelerating proteinsGDI: GDP Dissociation InhibitorGEF: Guanine nucleotide exchange factorHDAC: Histone deacetylaseLIMK: LIM KinaseMAL: Megakaryoblastic leukaemiaMARCKS: Myristoylated alanine-rich C-kinase substrateMKL: Megakaryoblastic leukaemiaMLC: Myosin light chainMRTF: Myocardin Related Transcription FactorOTT: One-Twenty Two ProteinPACSIN2: Protein Kinase C And Casein Kinase Substrate In Neurons 2PAK: P21-Activated KinasePDK: Pyruvate Dehydrogenase kinasePI3K: Phosphoinositide 3-kinasePKC: Protein kinase CPTPRJ: Protein tyrosine phosphatase receptor type JRAC: Ras-related C3 botulinum toxin substrate 1RBM15: RNA Binding Motif Protein 15RHO: Ras homologousROCK: Rho-associated protein kinaseSCAR: Suppressor of cAMP receptorSRF: Serum response factorSRC: SarcTAZ: Transcriptional coactivator with PDZ motifTUBB1: Tubulin β1VEGF: Vascular endothelial growth factorWAS: Wiskott Aldrich syndromeWASP: Wiskott Aldrich syndrome proteinWAVE: WASP-family verprolin-homologous proteinWIP: WASP-interacting proteinYAP: Yes-associated protein
Collapse
Affiliation(s)
- William Vainchenker
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France.,GrEX, Sorbonne Paris Cité, Paris, France
| | - Brahim Arkoun
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France.,GrEX, Sorbonne Paris Cité, Paris, France
| | - Francesca Basso-Valentina
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France.,Université Sorbonne Paris Cité/Université Paris Dideront, Paris, France
| | - Larissa Lordier
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France
| | - Najet Debili
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France
| | - Hana Raslova
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France
| |
Collapse
|
24
|
Zhou J, Yan Q, Tang C, Liao Y, Zhang Q, Wang X, Zhou X, Lai L, Zou Q. Development of a rabbit model of Wiskott-Aldrich syndrome. FASEB J 2020; 35:e21226. [PMID: 33236397 DOI: 10.1096/fj.202002118rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 11/11/2022]
Abstract
The Wiskott-Aldrich syndrome (WAS) is a severe recessive X-linked immunodeficiency resulting from loss-of-function mutations in the WAS gene. Mouse is the only mammalian model used for investigation of WAS pathogenesis. However, the mouse model does not accurately recapitulate WAS clinical phenotypes, thus, limiting its application in WAS clinical research. Herein, we report the generation of WAS knockout (KO) rabbits via embryo co-injection of Cas9 mRNA and a pair of sgRNAs targeting exons 2 and 7. WAS KO rabbits exhibited many symptoms similar to those of WAS patients, including thrombocytopenia, bleeding tendency, infections, and reduced numbers of T cell in the spleen and peripheral blood. The WAS KO rabbit model provides a new valuable tool for preclinical trials of WAS treatment.
Collapse
Affiliation(s)
- Juanjuan Zhou
- School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Quanmei Yan
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Chengcheng Tang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Yuan Liao
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Quanjun Zhang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China
| | - Xiaomin Wang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaoqing Zhou
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Liangxue Lai
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China
| | - Qingjian Zou
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| |
Collapse
|
25
|
Nicolai L, Schiefelbein K, Lipsky S, Leunig A, Hoffknecht M, Pekayvaz K, Raude B, Marx C, Ehrlich A, Pircher J, Zhang Z, Saleh I, Marel AK, Löf A, Petzold T, Lorenz M, Stark K, Pick R, Rosenberger G, Weckbach L, Uhl B, Xia S, Reichel CA, Walzog B, Schulz C, Zheden V, Bender M, Li R, Massberg S, Gaertner F. Vascular surveillance by haptotactic blood platelets in inflammation and infection. Nat Commun 2020; 11:5778. [PMID: 33188196 PMCID: PMC7666582 DOI: 10.1038/s41467-020-19515-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 10/14/2020] [Indexed: 12/21/2022] Open
Abstract
Breakdown of vascular barriers is a major complication of inflammatory diseases. Anucleate platelets form blood-clots during thrombosis, but also play a crucial role in inflammation. While spatio-temporal dynamics of clot formation are well characterized, the cell-biological mechanisms of platelet recruitment to inflammatory micro-environments remain incompletely understood. Here we identify Arp2/3-dependent lamellipodia formation as a prominent morphological feature of immune-responsive platelets. Platelets use lamellipodia to scan for fibrin(ogen) deposited on the inflamed vasculature and to directionally spread, to polarize and to govern haptotactic migration along gradients of the adhesive ligand. Platelet-specific abrogation of Arp2/3 interferes with haptotactic repositioning of platelets to microlesions, thus impairing vascular sealing and provoking inflammatory microbleeding. During infection, haptotaxis promotes capture of bacteria and prevents hematogenic dissemination, rendering platelets gate-keepers of the inflamed microvasculature. Consequently, these findings identify haptotaxis as a key effector function of immune-responsive platelets. Breakdown of vascular barriers is a major complication of inflammatory diseases. However, the mechanisms underlying platelet recruitment to inflammatory micro-environments remains unclear. Here, the authors identify haptotaxis as a key effector function of immune-responsive platelets
Collapse
Affiliation(s)
- Leo Nicolai
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802, Munich, Germany
| | - Karin Schiefelbein
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Silvia Lipsky
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Alexander Leunig
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Marie Hoffknecht
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Ben Raude
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Charlotte Marx
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Andreas Ehrlich
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Joachim Pircher
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Zhe Zhang
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Inas Saleh
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | | | - Achim Löf
- Ludwig-Maximilians-Universität, 80799, Munich, Germany
| | - Tobias Petzold
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Michael Lorenz
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Robert Pick
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, München, Germany.,Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Planegg-Martinsried, Munich, Germany
| | - Gerhild Rosenberger
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Ludwig Weckbach
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany
| | - Bernd Uhl
- Department of Otorhinolarynology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Sheng Xia
- Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA
| | | | - Barbara Walzog
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, München, Germany.,Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Planegg-Martinsried, Munich, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802, Munich, Germany
| | - Vanessa Zheden
- Institute of Science and Technology (IST) Austria, 3400, Klosterneuburg, Austria
| | - Markus Bender
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Rong Li
- Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität, 81377, Munich, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802, Munich, Germany.
| | - Florian Gaertner
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802, Munich, Germany. .,Institute of Science and Technology (IST) Austria, 3400, Klosterneuburg, Austria.
| |
Collapse
|
26
|
Muñoz P, Tristán-Manzano M, Sánchez-Gilabert A, Santilli G, Galy A, Thrasher AJ, Martin F. WAS Promoter-Driven Lentiviral Vectors Mimic Closely the Lopsided WASP Expression during Megakaryocytic Differentiation. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:220-235. [PMID: 33102615 PMCID: PMC7558809 DOI: 10.1016/j.omtm.2020.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/11/2020] [Indexed: 01/10/2023]
Abstract
Transplant of gene-modified autologous hematopoietic progenitors cells has emerged as a new therapeutic approach for Wiskott-Aldrich syndrome (WAS), a primary immunodeficiency with microthrombocytopenia and abnormal lymphoid and myeloid functions. Despite the clinical benefits obtained in ongoing clinical trials, platelet restoration is suboptimal. The incomplete restoration of platelets in these patients can be explained either by a low number of corrected cells or by insufficient or inadequate WASP expression during megakaryocyte differentiation and/or in platelets. We therefore used in vitro models to study the endogenous WASP expression pattern during megakaryocytic differentiation and compared it with the expression profiles achieved by different therapeutic lentiviral vectors (LVs) driving WAS cDNA through different regions of the WAS promoter. Our data showed that all WAS promoter-driven LVs mimic very closely the endogenous WAS expression kinetic during megakaryocytic differentiation. However, LVs harboring the full-length (1.6-kb) WAS-proximal promoter (WW1.6) or a combination of the WAS alternative and proximal promoters (named AW) had the best behavior. Finally, all WAS-driven LVs restored the WAS knockout (WASKO) mice phenotype and functional defects of hematopoietic stem and progenitor cells (HSPCs) from a WAS patient with similar efficiency. In summary, our data back up the use of WW1.6 and AW LVs as physiological gene transfer tools for WAS therapy.
Collapse
Affiliation(s)
- Pilar Muñoz
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Ilustracion 114, 18016 Granada, Spain.,University College London (UCL) Great Ormond Street Institute of Child Health (ICH), 30 Guilford Street, WC1N 1EH London, UK
| | - María Tristán-Manzano
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Ilustracion 114, 18016 Granada, Spain
| | - Almudena Sánchez-Gilabert
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Ilustracion 114, 18016 Granada, Spain
| | - Giorgia Santilli
- University College London (UCL) Great Ormond Street Institute of Child Health (ICH), 30 Guilford Street, WC1N 1EH London, UK
| | - Anne Galy
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951, 91000 Evry, France
| | - Adrian J Thrasher
- University College London (UCL) Great Ormond Street Institute of Child Health (ICH), 30 Guilford Street, WC1N 1EH London, UK
| | - Francisco Martin
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Ilustracion 114, 18016 Granada, Spain
| |
Collapse
|
27
|
Mansour R, El-Orfali Y, Saber A, Noun D, Youssef N, Youssef Y, Hanna-Wakim R, Dbaibo G, Abboud M, Massaad MJ. Wiskott-Aldrich Syndrome in four male siblings from a consanguineous family from Lebanon. Clin Immunol 2020; 219:108573. [PMID: 32814211 DOI: 10.1016/j.clim.2020.108573] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency disorder (PID) characterized by microthrombocytopenia, bloody diarrhea, eczema, recurrent infections, and a high incidence of autoimmunity and malignancy. OBJECTIVE To investigate the mechanism of thrombocytopenia and infections in four boys of consanguineous parents from Lebanon. METHODS Patient gDNA was studied using Next Generation Sequencing and Sanger Sequencing. Protein expression was determined by immunoblotting, and mRNA expression by semi-quantitative RT-PCR. F-actin polymerization and cellular proliferation were assayed by flow cytometry. RESULTS We identified a threonine to a methionine change at position 45 (T45M) of the WAS protein (WASp) that abolished protein expression and disturbed F-actin polymerization and T cell proliferation, but not B cell proliferation. In addition, the levels of the WAS-interacting protein (WIP) were significantly decreased in the patients. CONCLUSION The mutation identified severely destabilizes WASp and affects the downstream signaling events important for T cell function, but not B cell function. It was previously known that the stability of WASp depends on WIP. In this manuscript, we report that the stability of WIP also depends on WASp. Finally, it is important to suspect X-linked PIDs even in consanguineous families. CLINICAL IMPLICATIONS The patients are above the optimal age for transplant in WAS, and it is difficult to identify one or more donors for four patients, therefore, they represent ideal candidates for gene therapy or interleukin-2 therapy.
Collapse
Affiliation(s)
- Rana Mansour
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Youmna El-Orfali
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Antoine Saber
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Dolly Noun
- Division of Pediatric Hematology Oncology, Department of Pediatrics and Adolescent Medicine, Beirut, Lebanon; Children's Cancer Center of Lebanon, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nour Youssef
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yolla Youssef
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rima Hanna-Wakim
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Ghassan Dbaibo
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; Department of Biochemistry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Miguel Abboud
- Division of Pediatric Hematology Oncology, Department of Pediatrics and Adolescent Medicine, Beirut, Lebanon; Children's Cancer Center of Lebanon, American University of Beirut Medical Center, Beirut, Lebanon
| | - Michel J Massaad
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
28
|
Rai R, Romito M, Rivers E, Turchiano G, Blattner G, Vetharoy W, Ladon D, Andrieux G, Zhang F, Zinicola M, Leon-Rico D, Santilli G, Thrasher AJ, Cavazza A. Targeted gene correction of human hematopoietic stem cells for the treatment of Wiskott - Aldrich Syndrome. Nat Commun 2020; 11:4034. [PMID: 32788576 PMCID: PMC7423939 DOI: 10.1038/s41467-020-17626-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 07/03/2020] [Indexed: 12/31/2022] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is an X-linked primary immunodeficiency with severe platelet abnormalities and complex immunodeficiency. Although clinical gene therapy approaches using lentiviral vectors have produced encouraging results, full immune and platelet reconstitution is not always achieved. Here we show that a CRISPR/Cas9-based genome editing strategy allows the precise correction of WAS mutations in up to 60% of human hematopoietic stem and progenitor cells (HSPCs), without impairing cell viability and differentiation potential. Delivery of the editing reagents to WAS HSPCs led to full rescue of WASp expression and correction of functional defects in myeloid and lymphoid cells. Primary and secondary transplantation of corrected WAS HSPCs into immunodeficient mice showed persistence of edited cells for up to 26 weeks and efficient targeting of long-term repopulating stem cells. Finally, no major genotoxicity was associated with the gene editing process, paving the way for an alternative, yet highly efficient and safe therapy.
Collapse
Affiliation(s)
- Rajeev Rai
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Marianna Romito
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Elizabeth Rivers
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Giandomenico Turchiano
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Georges Blattner
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Winston Vetharoy
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Dariusz Ladon
- SIHMDS-Acquired Genomics, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and System Medicine, University of Freiburg, 26 Stefan-Meier-Strasse, 79104, Freiburg, Germany
| | - Fang Zhang
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Marta Zinicola
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Diego Leon-Rico
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Giorgia Santilli
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Adrian J Thrasher
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Alessia Cavazza
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
29
|
Blumenthal D, Chandra V, Avery L, Burkhardt JK. Mouse T cell priming is enhanced by maturation-dependent stiffening of the dendritic cell cortex. eLife 2020; 9:e55995. [PMID: 32720892 PMCID: PMC7417170 DOI: 10.7554/elife.55995] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/27/2020] [Indexed: 12/28/2022] Open
Abstract
T cell activation by dendritic cells (DCs) involves forces exerted by the T cell actin cytoskeleton, which are opposed by the cortical cytoskeleton of the interacting antigen-presenting cell. During an immune response, DCs undergo a maturation process that optimizes their ability to efficiently prime naïve T cells. Using atomic force microscopy, we find that during maturation, DC cortical stiffness increases via a process that involves actin polymerization. Using stimulatory hydrogels and DCs expressing mutant cytoskeletal proteins, we find that increasing stiffness lowers the agonist dose needed for T cell activation. CD4+ T cells exhibit much more profound stiffness dependency than CD8+ T cells. Finally, stiffness responses are most robust when T cells are stimulated with pMHC rather than anti-CD3ε, consistent with a mechanosensing mechanism involving receptor deformation. Taken together, our data reveal that maturation-associated cytoskeletal changes alter the biophysical properties of DCs, providing mechanical cues that costimulate T cell activation.
Collapse
Affiliation(s)
- Daniel Blumenthal
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Vidhi Chandra
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Lyndsay Avery
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
30
|
Saeed MB, Record J, Westerberg LS. Two sides of the coin: Cytoskeletal regulation of immune synapses in cancer and primary immune deficiencies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:1-97. [DOI: 10.1016/bs.ircmb.2020.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Du SW, Arkatkar T, Al Qureshah F, Jacobs HM, Thouvenel CD, Chiang K, Largent AD, Li QZ, Hou B, Rawlings DJ, Jackson SW. Functional Characterization of CD11c + Age-Associated B Cells as Memory B Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:2817-2826. [PMID: 31636237 DOI: 10.4049/jimmunol.1900404] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/24/2019] [Indexed: 12/25/2022]
Abstract
Age-associated B cells (ABCs) are a unique subset of B cells defined by surface CD11b and CD11c expression. Although ABC expansion has been observed in both human and animal studies in the setting of advanced age, during humoral autoimmunity and following viral infection, the functional properties of this cellular subset remain incompletely defined. In the current study, we demonstrate that ABCs fulfill the criteria for memory B cells (MBCs), based on evidence of Ag-dependent expansion and persistence in a state poised for rapid differentiation into Ab-secreting plasma cells during secondary responses. First, we show that a majority of ABCs are not actively cycling but exhibit an extensive replication history consistent with prior Ag engagement. Second, despite unswitched surface IgM expression, ABCs show evidence of activation-induced cytidine deaminase (AID)-dependent somatic hypermutation. Third, BCRs cloned from sorted ABCs exhibit broad autoreactivity and polyreactivity. Although the overall level of ABC self-reactivity was not increased relative to naive B cells, ABCs lacked features of functional anergy characteristic of autoreactive B cells. Fourth, ABCs express MBC surface markers consistent with being poised for rapid plasma cell differentiation during recall responses. Finally, in a murine model of viral infection, adoptively transferred CD11c+ B cells rapidly differentiated into class-switched Ab-secreting cells upon Ag rechallenge. In summary, we phenotypically and functionally characterize ABCs as IgM-expressing MBCs, findings that together implicate ABCs in the pathogenesis of systemic autoimmunity.
Collapse
Affiliation(s)
- Samuel W Du
- Seattle Children's Research Institute, Seattle, WA 98101
| | - Tanvi Arkatkar
- Seattle Children's Research Institute, Seattle, WA 98101
| | - Fahd Al Qureshah
- Seattle Children's Research Institute, Seattle, WA 98101.,King Abdulaziz City for Science and Technology, Riyadh 12354, Saudi Arabia.,Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109
| | - Holly M Jacobs
- Seattle Children's Research Institute, Seattle, WA 98101
| | | | - Kristy Chiang
- Seattle Children's Research Institute, Seattle, WA 98101
| | | | - Quan-Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Baidong Hou
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and
| | - David J Rawlings
- Seattle Children's Research Institute, Seattle, WA 98101.,Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195
| | - Shaun W Jackson
- Seattle Children's Research Institute, Seattle, WA 98101; .,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195
| |
Collapse
|
32
|
Batcha MM, Ahamed AS, Peng CF. Identification of a new type of haematopoietic progenitor kinase-interacting protein (HIP-55) in Aedes aegypti mosquito haemocytes and its involvement in immunity-like functions in mosquito: a molecular study. Parasitol Res 2019; 118:2509-2521. [PMID: 31377908 DOI: 10.1007/s00436-019-06408-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/25/2019] [Indexed: 11/26/2022]
Abstract
In this study, we characterize the HIP-55 protein in the mosquito Aedes aegypti for the first time. HIP-55 is a 55-kDa HPK1-interacting protein that is also called SH3P7. HIP-55 constitutively binds HPK1 'via' an HPK1 proline-rich motif 2(PR2) through its C-terminal SH3 domain. HIP-55 critically interacts with ZAP-70, and this interaction was induced by TCR signalling. ZAP-70 phosphorylated HIP-55 at Tyr-334 and Tyr-344 in vitro and in vivo. In our previous findings, AaZAP gene expression strongly proved that AaZAP-70 was involved in immunity-like functions in mosquito. Northern blot analysis of HIP-55 mRNA expression confirmed that it is only expressed in the abdomen and haemocyte tissues; this prediction correlates 100% and a polyclonal antibody also confirmed its localization in haemocytes and the abdomen. We prepared extracts to show the cytoplasmic expression (CE) of this protein. Previous results had proven that this protein is secreted from the cytoplasm; thus, we confirmed here that the protein is a cytoplasmic adaptor protein in mosquitoes and mammalian systems. Furthermore, our polyclonal antibody against HIP-55 also demonstrated that this protein is found in haemocytes and abdomen tissues, which assumes that the protein may be involved in phagocytic-like functions. RNAi (siRNA) silencing studies were used to degrade mosquito HIP-55; however, silencing only slightly affected the HIP-55 sequence and the gene transcriptional level. To characterize this protein, we cloned 609 bp from the 1.6-kb full-length cDNA using a pET28 vector for polyclonal antibody production. Graphical abstract.
Collapse
Affiliation(s)
- M Mohiadeen Batcha
- Department of Zoology, HKRH College, Uthamapalayam, Theni District, Tamil Nadu, India.
- Post Doctoral Scientist, Institute of Tropical Medicine, National Yang Ming University, Shihpai, Taipei-112, Taiwan, Republic of China.
| | - A Sajith Ahamed
- Department of Microbiology, HKRH College, Uthamapalayam, Theni District, Tamil Nadu, India
| | - Chiung Fang Peng
- Fuga Biotechnology, Chongqing S. Rd, Zhongzheng Dist, Taipei - 100, Taiwan
| |
Collapse
|
33
|
Jin YY, Wu J, Chen TX, Chen J. When WAS Gene Diagnosis Is Needed: Seeking Clues Through Comparison Between Patients With Wiskott-Aldrich Syndrome and Idiopathic Thrombocytopenic Purpura. Front Immunol 2019; 10:1549. [PMID: 31354712 PMCID: PMC6634258 DOI: 10.3389/fimmu.2019.01549] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/21/2019] [Indexed: 12/31/2022] Open
Abstract
Background: Wiskott-Aldrich syndrome (WAS) is a rare and severe X-linked disorder with variable clinical phenotypes correlating with the type of mutations in the WAS gene. The syndrome is difficult to differentiate from idiopathic thrombocytopenic purpura (ITP) before genetic diagnosis. We retrospectively reviewed patients suspected to have WAS who were referred to our hospital from 2004 to 2016 and compared the clinical features and laboratory examination of genetically confirmed WAS patients and of patients diagnosed with ITP in order to seek some clues to distinguish WAS and ITP before genetic diagnosis. Methods: Seventy-eight children suspected to have WAS from 78 unrelated families were enrolled in this study. The clinical data and laboratory examination of children were reviewed in the present study. The distribution of lymphocyte subsets from peripheral blood was examined by how cytometry. WASP mutations were identified by direct sequencing of PCR-amplified genomic DNA. Results: Forty-two patients were finally diagnosed with WAS genetically. The median onset age of these patients was 1 month (range: 1 day−10 months). The median diagnosis lag was 4.6 months (range: 0 months−9.42 years). Fifteen patients (35.71%) had positive family histories. More than half of the patients (n = 23, 54.76%) had diarrhea. Twenty-three (54.76%) had pneumonia, 7 with severe symptoms. Major bleeding events included skin spots or petechiae (n = 27, 64.29%), per-rectal bleeding (n = 21, 50.00%), epistaxis (n = 7, 16.67%) and intracranial bleeding (n = 2, 4.76%). Twenty-nine patients (69.05%) had eczema, and one patient had a drug allergy. Three patients had autoimmune diseases, among whom 2 had autoimmune hemolytic anemia and one had autoimmune hemolytic anemia and IgA nephropathy. A total of 42 mutations in WASP were identified, including 19 novel mutations. Eight patients received hematopoietic stem cell transplantation (HSCT) and all survived. Compared with the 30 patients diagnosed with ITP, the WAS patients had higher EOS counts and elevated IgE level, increased NK cell numbers but fewer CD8+T lymphocytes. Conclusion: The WAS gene diagnosis should be considered in all males with ITP-like features, especially for patients with a very early onset age, decreased MPV (<6.5 fl), higher EOS counts and elevated IgE level, increased NK cell number, diminished CD8+T lymphocyte count.
Collapse
Affiliation(s)
- Ying-Ying Jin
- Department of Rheumatology/Immunology, Children's National Medical Center, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wu
- Division of Immunology, Institute of Pediatric Translational Medicine, Children's National Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong-Xin Chen
- Department of Rheumatology/Immunology, Children's National Medical Center, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Division of Immunology, Institute of Pediatric Translational Medicine, Children's National Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji Chen
- Department of Dermatology, Children's National Medical Center, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Sun X, Wei Y, Lee PP, Ren B, Liu C. The role of WASp in T cells and B cells. Cell Immunol 2019; 341:103919. [PMID: 31047647 DOI: 10.1016/j.cellimm.2019.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/08/2019] [Accepted: 04/16/2019] [Indexed: 12/21/2022]
Abstract
Wiskott-Aldrich syndrome (WAS) is a form of primary immunodeficiency (PIDs) resulting from mutations of the gene that encodes Wiskott-Aldrich syndrome protein (WASp). WASp is the first identified and most widely studied protein belonging to the actin nucleation-promoting factor family and plays significant role in integrating and transforming signals from critical receptors on the cell surface to actin remodeling. WASp functions in immune defense and homeostasis through the regulation of actin cytoskeleton-dependent cellular processes as well as processes uncoupled with actin polymerization like nuclear transcription programs. In this article, we review the mechanisms of WASp activation through an understanding of its structure. We further discuss the role of WASp in adaptive immunity, paying special attention to some recent findings on the crucial role of WASp in the formation of immunological synapse, the regulation of T follicular helper (Tfh) cells and in the prevention of autoimmunity.
Collapse
Affiliation(s)
- Xizi Sun
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yin Wei
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Pamela P Lee
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Boxu Ren
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China.
| | - Chaohong Liu
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.
| |
Collapse
|
35
|
Baptista MAP, Westerberg LS. Activation of compensatory pathways via Rac2 in the absence of the Cdc42 effector Wiskott-Aldrich syndrome protein in Dendritic cells. Small GTPases 2019; 10:81-88. [PMID: 28129089 PMCID: PMC6380290 DOI: 10.1080/21541248.2016.1275363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 10/20/2022] Open
Abstract
There is extensive crosstalk between different Rho GTPases, including Cdc42, Rac1, and Rac2, and they can activate or inhibit the activity of each other. Dendritic cells express both Rac1 and Rac2. Due to posttranslational modification of lipid anchors, Rac1 localizes mainly to the plasma membrane whereas Rac2 localizes to the phagosomal membrane where it assembles the NADPH complex. Our recent study of primary immunodeficiency disease caused by mutations in the Cdc42 effector Wiskott-Aldrich syndrome protein (WASp) has shed light on the compensatory mechanisms between Rho GTPases and their effector proteins. WASp-deficient dendritic cells have increased localization and activity of Rac2 to the phagosomal membrane and this allows antigen to be presented on MHC class I molecules to activate cytotoxic CD8+ T cells. This study reveals an intricate balance between Rac2 and WASp signaling pathways and provides an example of compensatory pathways in cells devoid of the Cdc42 effector WASp.
Collapse
Affiliation(s)
- Marisa A. P. Baptista
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Lisa S. Westerberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
|
37
|
Pazmandi J, Kalinichenko A, Ardy RC, Boztug K. Early-onset inflammatory bowel disease as a model disease to identify key regulators of immune homeostasis mechanisms. Immunol Rev 2019; 287:162-185. [PMID: 30565237 PMCID: PMC7379380 DOI: 10.1111/imr.12726] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 09/23/2018] [Indexed: 12/11/2022]
Abstract
Rare, monogenetic diseases present unique models to dissect gene functions and biological pathways, concomitantly enhancing our understanding of the etiology of complex (and often more common) traits. Although inflammatory bowel disease (IBD) is a generally prototypic complex disease, it can also manifest in an early-onset, monogenic fashion, often following Mendelian modes of inheritance. Recent advances in genomic technologies have spurred the identification of genetic defects underlying rare, very early-onset IBD (VEO-IBD) as a disease subgroup driven by strong genetic influence, pinpointing key players in the delicate homeostasis of the immune system in the gut and illustrating the intimate relationships between bowel inflammation, systemic immune dysregulation, and primary immunodeficiency with increased susceptibility to infections. As for other human diseases, it is likely that adult-onset diseases may represent complex diseases integrating the effects of host genetic susceptibility and environmental triggers. Comparison of adult-onset IBD and VEO-IBD thus provides beautiful models to investigate the relationship between monogenic and multifactorial/polygenic diseases. This review discusses the present and novel findings regarding monogenic IBD as well as key questions and future directions of IBD research.
Collapse
Affiliation(s)
- Julia Pazmandi
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Artem Kalinichenko
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Rico Chandra Ardy
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
- Department of PediatricsSt. Anna Kinderspital and Children's Cancer Research InstituteMedical University of ViennaViennaAustria
| |
Collapse
|
38
|
Wiskott-Aldrich syndrome protein (WASP) is a tumor suppressor in T cell lymphoma. Nat Med 2018; 25:130-140. [PMID: 30510251 DOI: 10.1038/s41591-018-0262-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 09/25/2018] [Indexed: 01/10/2023]
Abstract
In T lymphocytes, the Wiskott-Aldrich Syndrome protein (WASP) and WASP-interacting-protein (WIP) regulate T cell antigen receptor (TCR) signaling, but their role in lymphoma is largely unknown. Here we show that the expression of WASP and WIP is frequently low or absent in anaplastic large cell lymphoma (ALCL) compared to other T cell lymphomas. In anaplastic lymphoma kinase-positive (ALK+) ALCL, WASP and WIP expression is regulated by ALK oncogenic activity via its downstream mediators STAT3 and C/EBP-β. ALK+ lymphomas were accelerated in WASP- and WIP-deficient mice. In the absence of WASP, active GTP-bound CDC42 was increased and the genetic deletion of one CDC42 allele was sufficient to impair lymphoma growth. WASP-deficient lymphoma showed increased mitogen-activated protein kinase (MAPK) pathway activation that could be exploited as a therapeutic vulnerability. Our findings demonstrate that WASP and WIP are tumor suppressors in T cell lymphoma and suggest that MAP-kinase kinase (MEK) inhibitors combined with ALK inhibitors could achieve a more potent therapeutic effect in ALK+ ALCL.
Collapse
|
39
|
Tang R, Langdon WY, Zhang J. Regulation of immune responses by E3 ubiquitin ligase Cbl-b. Cell Immunol 2018; 340:103878. [PMID: 30442330 DOI: 10.1016/j.cellimm.2018.11.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/04/2018] [Accepted: 11/05/2018] [Indexed: 12/21/2022]
Abstract
Casitas B lymphoma-b (Cbl-b), a RING finger E3 ubiquitin ligase, has been identified as a critical regulator of adaptive immune responses. Cbl-b is essential for establishing the threshold for T cell activation and regulating peripheral T cell tolerance through various mechanisms. Intriguingly, recent studies indicate that Cbl-b also modulates innate immune responses, and plays a key role in host defense to pathogens and anti-tumor immunity. These studies suggest that targeting Cbl-b may represent a potential therapeutic strategy for the management of human immune-related disorders such as autoimmune diseases, infections, tumors, and allergic airway inflammation. In this review, we summarize the latest developments regarding the roles of Cbl-b in innate and adaptive immunity, and immune-mediated diseases.
Collapse
Affiliation(s)
- Rong Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Wallace Y Langdon
- School of Biological Sciences, University of Western Australia, Perth, Australia
| | - Jian Zhang
- Department of Pathology, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
40
|
Du SW, Arkatkar T, Jacobs HM, Rawlings DJ, Jackson SW. Generation of functional murine CD11c + age-associated B cells in the absence of B cell T-bet expression. Eur J Immunol 2018; 49:170-178. [PMID: 30353919 DOI: 10.1002/eji.201847641] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/21/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
Age-associated B cells (ABC), a novel subset of activated B cells defined by CD11b and CD11c expression, have been linked with both protective anti-viral responses and the pathogenesis of systemic autoimmunity. Expression of the TH 1 lineage transcription factor T-bet has been identified as a defining feature of ABC biology, with B cell-intrinsic expression of this transcription factor proposed to be required for ABC formation. In contrast to this model, we report that Tbx21 (encoding T-bet)-deficient B cells upregulate CD11b and CD11c surface expression in vitro in response to integrated TLR and cytokine signals. Moreover, B cell-intrinsic T-bet deletion in a murine lupus model exerted no impact of ABC generation in vivo, with Tbx21-/- ABCs exhibiting an identical surface phenotype to wild-type (WT) ABCs. Importantly, WT and Tbx21-/- ABCs sorted from autoimmune mice produced equivalent amounts of IgM and IgG ex vivo following TLR stimulation, indicating that T-bet-deficient ABCs are likely functional in vivo. In summary, our data contradict the established literature by demonstrating that T-bet expression is not uniformly required for ABC generation.
Collapse
Affiliation(s)
- Samuel W Du
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA
| | - Tanvi Arkatkar
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA
| | - Holly M Jacobs
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.,Departments of Immunology and Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Shaun W Jackson
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA.,Departments of Immunology and Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
41
|
Schwinger W, Urban C, Ulreich R, Sperl D, Karastaneva A, Strenger V, Lackner H, Boztug K, Albert MH, Benesch M, Seidel MG. The Phenotype and Treatment of WIP Deficiency: Literature Synopsis and Review of a Patient With Pre-transplant Serial Donor Lymphocyte Infusions to Eliminate CMV. Front Immunol 2018; 9:2554. [PMID: 30450104 PMCID: PMC6224452 DOI: 10.3389/fimmu.2018.02554] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/17/2018] [Indexed: 11/13/2022] Open
Abstract
Early diagnosis of primary immunodeficiency disorders (PID) is vital and allows directed treatment, especially in syndromes with severe or profound combined immunodeficiency. In PID patients with perinatal CMV or other opportunistic, invasive infections (e.g., Pneumocystis or Aspergillus), multi-organ morbidity may already arise within the first months of life, before hematopoietic stem cell transplantation (HSCT) or gene therapy can be undertaken, compromising the definitive treatment and outcome. Deficiency of Wiskott-Aldrich syndrome (WAS) protein-interacting protein (WIP deficiency) causes an autosomal recessive, WAS-like syndrome with early-onset combined immunodeficiency that has been described in three pedigrees to date. While WAS typically includes combined immunodeficiency, microthrombocytopenia, and eczema, the clinical and laboratory phenotypes of WIP-deficient patients-including lymphocyte subsets, platelets, lymphocyte proliferation in vitro, and IgE-varied widely and did not entirely recapitulate WAS, impeding early diagnosis in the reported patients. To elucidate the phenotype of WIP deficiency, we provide a comprehensive synopsis of clinical and laboratory features of all hitherto-described patients (n = 6) and WIP negative mice. Furthermore, we summarize the treatment modalities and outcomes of these patients and review in detail the course of one of them who was successfully treated with serial, unconditioned, maternal, HLA-identical donor lymphocyte infusions (DLI) against life-threatening, invasive CMV infection, followed by a TCRαβ/CD19-depleted, treosulfan/melphalan-conditioned, peripheral blood HSCT and repetitive, secondary-prophylactic, CMV-specific DLI with 1-year post-HSCT follow-up. This strategy could be useful in other patients with substantial premorbidity, considered "too bad to transplant," who have an HLA-identical family donor, to eliminate infections and bridge until definitive treatment.
Collapse
Affiliation(s)
- Wolfgang Schwinger
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Christian Urban
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Raphael Ulreich
- Pediatric Intensive Care Unit, Division of General Pediatrics, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Daniela Sperl
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Anna Karastaneva
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Volker Strenger
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Herwig Lackner
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Department of Pediatrics, St. Anna Kinderspital and Children's Cancer Research Institute, Medical University of Vienna, Vienna, Austria
| | - Michael H Albert
- Dr. von Hauner University Children's Hospital, Ludwig Maximilian Universität, Munich, Germany
| | - Martin Benesch
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Markus G Seidel
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria.,Research Unit for Pediatric Hematology and Immunology, Medical University Graz, Graz, Austria
| |
Collapse
|
42
|
Cervantes-Luevano KE, Caronni N, Castiello MC, Fontana E, Piperno GM, Naseem A, Uva P, Bosticardo M, Marcovecchio GE, Notarangelo LD, Cicalese MP, Aiuti A, Villa A, Benvenuti F. Neutrophils drive type I interferon production and autoantibodies in patients with Wiskott-Aldrich syndrome. J Allergy Clin Immunol 2018; 142:1605-1617.e4. [PMID: 29447842 PMCID: PMC6089666 DOI: 10.1016/j.jaci.2017.11.063] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/27/2017] [Accepted: 11/21/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Wiskott-Aldrich syndrome (WAS) is a rare primary immunodeficiency caused by mutations in Wiskott-Aldrich syndrome protein (WASp), a key regulator of cytoskeletal dynamics in hematopoietic cells. A high proportion of patients experience autoimmunity caused by a breakdown in T- and B-cell tolerance. Moreover, excessive production of type I interferon (IFN-I) by plasmacytoid dendritic cells (pDCs) contributes to autoimmune signs; however, the factors that trigger excessive innate activation have not been defined. OBJECTIVE Neutrophil extracellular traps (NETs) emerged as major initiating factors in patients with diseases such as systemic lupus erythematosus and rheumatoid arthritis. In this study we explored the possible involvement of aberrant neutrophil functions in patients with WAS. METHODS We evaluated the expression of a set of granulocyte genes associated with NETs in a cohort of patients with WAS and the presence of NET inducers in sera. Using a mouse model of WAS, we analyzed NET release by WASp-null neutrophils and evaluated the composition and homeostasis of neutrophils in vivo. By using depletion experiments, we assessed the effect of neutrophils in promoting inflammation and reactivity against autoantigens. RESULTS Transcripts of genes encoding neutrophil enzymes and antimicrobial peptides were increased in granulocytes of patients with WAS, and serum-soluble factors triggered NET release. WASp-null neutrophils showed increased spontaneous NETosis, induced IFN-I production by pDCs, and activated B cells through B-cell activating factor. Consistently, their depletion abolished constitutive pDC activation, normalized circulating IFN-I levels, and, importantly, abolished production of autoantibodies directed against double-stranded DNA, nucleosomes, and myeloperoxidase. CONCLUSIONS These findings reveal that neutrophils are involved in the pathogenic loop that causes excessive activation of innate cells and autoreactive B cells, thus identifying novel mechanisms that contribute to the autoimmunity of WAS.
Collapse
Affiliation(s)
| | - Nicoletta Caronni
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Maria C Castiello
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Elena Fontana
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Giulia M Piperno
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Asma Naseem
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Paolo Uva
- CRS4, Science and Technology Park Polaris, Pula, Italy
| | - Marita Bosticardo
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy; Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Genni E Marcovecchio
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Maria P Cicalese
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy; San Raffaele Telethon Institute for Gene Therapy and the Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy; San Raffaele Telethon Institute for Gene Therapy and the Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy; Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Federica Benvenuti
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy.
| |
Collapse
|
43
|
Sawai CM, Serpas L, Neto AG, Jang G, Rashidfarrokhi A, Kolbeck R, Sanjuan MA, Reizis B, Sisirak V. Plasmacytoid Dendritic Cells Are Largely Dispensable for the Pathogenesis of Experimental Inflammatory Bowel Disease. Front Immunol 2018; 9:2475. [PMID: 30410494 PMCID: PMC6209677 DOI: 10.3389/fimmu.2018.02475] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/08/2018] [Indexed: 12/31/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition caused by an aberrant immune response to microbial components of the gastrointestinal tract. Plasmacytoid dendritic cells (pDCs) are innate immune cells specialized in the production of type I interferons and were recently implicated in the pathogenesis of autoimmune disorders such as lupus and scleroderma. While pDCs were shown to infiltrate intestinal mucosa of IBD patients and proposed to participate in intestinal inflammation, their net contribution to the disease remains unclear. We addressed this question by targeting the pDC-specific transcription factor TCF4 (E2-2) in experimental IBD caused by deficiency of Wiskott-Aldrich syndrome protein (WASP) or of interleukin-10 (IL-10). Monoallelic Tcf4 deletion, which was previously shown to abrogate experimental lupus, did not affect autoimmunity manifestations or colitis in WASP-deficient animals. Furthermore, conditional biallelic Tcf4 targeting resulted in a near-complete pDC ablation, yet had no effect on the development of colitis in IL-10-deficient mice. Our results suggest that, in contrast to other inflammatory and autoimmune diseases, pDCs do not play a major role in the pathogenesis of intestinal inflammation during IBD.
Collapse
Affiliation(s)
- Catherine M Sawai
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,INSERM, ACTION Laboratory, University of Bordeaux, Bordeaux, France
| | - Lee Serpas
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Antonio Galvao Neto
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Geunhyo Jang
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Roland Kolbeck
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD, United States
| | - Miguel A Sanjuan
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD, United States
| | - Boris Reizis
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Vanja Sisirak
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,CNRS-UMR, Immunoconcept, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
44
|
Ryzhakov G, West NR, Franchini F, Clare S, Ilott NE, Sansom SN, Bullers SJ, Pearson C, Costain A, Vaughan-Jackson A, Goettel JA, Ermann J, Horwitz BH, Buti L, Lu X, Mukhopadhyay S, Snapper SB, Powrie F. Alpha kinase 1 controls intestinal inflammation by suppressing the IL-12/Th1 axis. Nat Commun 2018; 9:3797. [PMID: 30228258 PMCID: PMC6143560 DOI: 10.1038/s41467-018-06085-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) are heterogenous disorders of the gastrointestinal tract caused by a spectrum of genetic and environmental factors. In mice, overlapping regions of chromosome 3 have been associated with susceptibility to IBD-like pathology, including a locus called Hiccs. However, the specific gene that controls disease susceptibility remains unknown. Here we identify a Hiccs locus gene, Alpk1 (encoding alpha kinase 1), as a potent regulator of intestinal inflammation. In response to infection with the commensal pathobiont Helicobacter hepaticus (Hh), Alpk1-deficient mice display exacerbated interleukin (IL)-12/IL-23 dependent colitis characterized by an enhanced Th1/interferon(IFN)-γ response. Alpk1 controls intestinal immunity via the hematopoietic system and is highly expressed by mononuclear phagocytes. In response to Hh, Alpk1-/- macrophages produce abnormally high amounts of IL-12, but not IL-23. This study demonstrates that Alpk1 promotes intestinal homoeostasis by regulating the balance of type 1/type 17 immunity following microbial challenge.
Collapse
Affiliation(s)
- Grigory Ryzhakov
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom
| | - Nathaniel R West
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom
- Genentech, Department of Cancer Immunology, South San Francisco, CA, 94080, USA
| | - Fanny Franchini
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom
| | - Simon Clare
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Nicholas E Ilott
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom
| | - Stephen N Sansom
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom
| | - Samuel J Bullers
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom
| | - Claire Pearson
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom
| | - Alice Costain
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom
| | - Alun Vaughan-Jackson
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom
| | - Jeremy A Goettel
- Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Joerg Ermann
- Department of Gastroenterology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Bruce H Horwitz
- Department of Gastroenterology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Ludovico Buti
- Ludwig Institute of Cancer Research, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Xin Lu
- Ludwig Institute of Cancer Research, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | | | - Scott B Snapper
- Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, United Kingdom.
| |
Collapse
|
45
|
Keszei M, Record J, Kritikou JS, Wurzer H, Geyer C, Thiemann M, Drescher P, Brauner H, Köcher L, James J, He M, Baptista MA, Dahlberg CI, Biswas A, Lain S, Lane DP, Song W, Pütsep K, Vandenberghe P, Snapper SB, Westerberg LS. Constitutive activation of WASp in X-linked neutropenia renders neutrophils hyperactive. J Clin Invest 2018; 128:4115-4131. [PMID: 30124469 PMCID: PMC6118594 DOI: 10.1172/jci64772] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023] Open
Abstract
Congenital neutropenia is characterized by low absolute neutrophil numbers in blood, leading to recurrent bacterial infections, and patients often require life-long granulocyte CSF (G-CSF) support. X-linked neutropenia (XLN) is caused by gain-of-function mutations in the actin regulator Wiskott-Aldrich syndrome protein (WASp). To understand the pathophysiology in XLN and the role of WASp in neutrophils, we here examined XLN patients and 2 XLN mouse models. XLN patients had reduced myelopoiesis and extremely low blood neutrophil number. However, their neutrophils had a hyperactive phenotype and were present in normal numbers in XLN patient saliva. Murine XLN neutrophils were hyperactivated, with increased actin dynamics and migration into tissues. We provide molecular evidence that the hyperactivity of XLN neutrophils is caused by WASp in a constitutively open conformation due to contingent phosphorylation of the critical tyrosine-293 and plasma membrane localization. This renders WASp activity less dependent on regulation by PI3K. Our data show that the amplitude of WASp activity inside a cell could be enhanced by cell-surface receptor signaling even in the context in which WASp is already in an active conformation. Moreover, these data categorize XLN as an atypical congenital neutropenia in which constitutive activation of WASp in tissue neutrophils compensates for reduced myelopoiesis.
Collapse
Affiliation(s)
- Marton Keszei
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Julien Record
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Joanna S. Kritikou
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hannah Wurzer
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Chiara Geyer
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Meike Thiemann
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Paul Drescher
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Brauner
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Laura Köcher
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jaime James
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Minghui He
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marisa A.P. Baptista
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Carin I.M. Dahlberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Amlan Biswas
- Gastroenterology Division, Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sonia Lain
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - David P. Lane
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wenxia Song
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Katrin Pütsep
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Vandenberghe
- Center for Human Genetics, Katholieke Universiteit (KU) Leuven and Hematology/Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Scott B. Snapper
- Gastroenterology Division, Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lisa S. Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Keppler SJ, Burbage M, Gasparrini F, Hartjes L, Aggarwal S, Massaad MJ, Geha RS, Bruckbauer A, Batista FD. The Lack of WIP Binding to Actin Results in Impaired B Cell Migration and Altered Humoral Immune Responses. Cell Rep 2018; 24:619-629. [PMID: 30021160 PMCID: PMC6077251 DOI: 10.1016/j.celrep.2018.06.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/11/2018] [Accepted: 06/12/2018] [Indexed: 11/27/2022] Open
Abstract
Wiskott-Aldrich syndrome protein (WASp) is a main cytoskeletal regulator in B cells. WASp-interacting protein (WIP) binds to and stabilizes WASp but also interacts with actin. Using mice with a mutated actin binding domain of WIP (WIPΔABD), we here investigated the role of WIP binding to actin during B cell activation. We found an altered differentiation of WIPΔABD B cells and diminished antibody affinity maturation after immunization. Mechanistically, WIPΔABD B cells showed impaired B cell receptor (BCR)-induced PI3K signaling and actin reorganization, likely caused by diminished CD81 expression and altered CD19 dynamics on the B cell surface. WIPΔABD B cells displayed reduced in vivo motility, concomitantly with impaired chemotaxis and defective F-actin polarization, HS1 phosphorylation, and polarization of HS1 to F-actin-rich structures after CXCL12 stimulation in vitro. We thus concluded that WIP binding to actin, independent of its binding to WASp, is critical for actin cytoskeleton plasticity in B cells.
Collapse
Affiliation(s)
- Selina Jessica Keppler
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, UK; MRI, TranslaTUM, Institute for Clinical Chemistry and Pathobiochemistry, Immune Signals and Cancer, 81675 Munich, Germany.
| | - Marianne Burbage
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, UK
| | | | - Lara Hartjes
- MRI, TranslaTUM, Institute for Clinical Chemistry and Pathobiochemistry, Immune Signals and Cancer, 81675 Munich, Germany
| | - Shweta Aggarwal
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, UK
| | - Michel J Massaad
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Raif S Geha
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Andreas Bruckbauer
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, UK
| | - Facundo D Batista
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, UK; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
47
|
The actin remodeling protein cofilin is crucial for thymic αβ but not γδ T-cell development. PLoS Biol 2018; 16:e2005380. [PMID: 29985916 PMCID: PMC6053251 DOI: 10.1371/journal.pbio.2005380] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 07/19/2018] [Accepted: 06/22/2018] [Indexed: 12/16/2022] Open
Abstract
Cofilin is an essential actin remodeling protein promoting depolymerization and severing of actin filaments. To address the relevance of cofilin for the development and function of T cells in vivo, we generated knock-in mice in which T-cell-specific nonfunctional (nf) cofilin was expressed instead of wild-type (WT) cofilin. Nf cofilin mice lacked peripheral αβ T cells and showed a severe thymus atrophy. This was caused by an early developmental arrest of thymocytes at the double negative (DN) stage. Importantly, even though DN thymocytes expressed the TCRβ chain intracellularly, they completely lacked TCRβ surface expression. In contrast, nf cofilin mice possessed normal numbers of γδ T cells. Their functionality was confirmed in the γδ T-cell-driven, imiquimod (IMQ)-induced, psoriasis-like murine model. Overall, this study not only highlights the importance of cofilin for early αβ T-cell development but also shows for the first time that an actin-binding protein is differentially involved in αβ versus γδ T-cell development.
Collapse
|
48
|
Pleines I, Cherpokova D, Bender M. Rho GTPases and their downstream effectors in megakaryocyte biology. Platelets 2018; 30:9-16. [DOI: 10.1080/09537104.2018.1478071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Irina Pleines
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Deya Cherpokova
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Markus Bender
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
49
|
Rodriguez Cetina Biefer H, Heinbokel T, Uehara H, Camacho V, Minami K, Nian Y, Koduru S, El Fatimy R, Ghiran I, Trachtenberg AJ, de la Fuente MA, Azuma H, Akbari O, Tullius SG, Vasudevan A, Elkhal A. Mast cells regulate CD4 + T-cell differentiation in the absence of antigen presentation. J Allergy Clin Immunol 2018; 142:1894-1908.e7. [PMID: 29470999 DOI: 10.1016/j.jaci.2018.01.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/19/2017] [Accepted: 01/28/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND Given their unique capacity for antigen uptake, processing, and presentation, antigen-presenting cells (APCs) are critical for initiating and regulating innate and adaptive immune responses. We have previously shown the role of nicotinamide adenine dinucleotide (NAD+) in T-cell differentiation independently of the cytokine milieu, whereas the precise mechanisms remained unknown. OBJECTIVE The objective of this study is to further dissect the mechanism of actions of NAD+ and determine the effect of APCs on NAD+-mediated T-cell activation. METHODS Isolated dendritic cells and bone marrow-derived mast cells (MCs) were used to characterize the mechanisms of action of NAD+ on CD4+ T-cell fate in vitro. Furthermore, NAD+-mediated CD4+ T-cell differentiation was investigated in vivo by using wild-type C57BL/6, MC-/-, MHC class II-/-, Wiskott-Aldrich syndrome protein (WASP)-/-, 5C.C7 recombination-activating gene 2 (Rag2)-/-, and CD11b-DTR transgenic mice. Finally, we tested the physiologic effect of NAD+ on the systemic immune response in the context of Listeria monocytogenes infection. RESULTS Our in vivo and in vitro findings indicate that after NAD+ administration, MCs exclusively promote CD4+ T-cell differentiation, both in the absence of antigen and independently of major APCs. Moreover, we found that MCs mediated CD4+ T-cell differentiation independently of MHC II and T-cell receptor signaling machinery. More importantly, although treatment with NAD+ resulted in decreased MHC II expression on CD11c+ cells, MC-mediated CD4+ T-cell differentiation rendered mice resistant to administration of lethal doses of L monocytogenes. CONCLUSIONS Collectively, our study unravels a novel cellular and molecular pathway that regulates innate and adaptive immunity through MCs exclusively and underscores the therapeutic potential of NAD+ in the context of primary immunodeficiencies and antimicrobial resistance.
Collapse
Affiliation(s)
- Hector Rodriguez Cetina Biefer
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Clinic for Cardiovascular Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Timm Heinbokel
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Department of Nephrology, Charite Universitaetsmedizin Berlin, Berlin, Germany
| | | | - Virginia Camacho
- Flow Cytometry Core Facility, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Boston, Mass
| | - Koichiro Minami
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Yeqi Nian
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Suresh Koduru
- School of Medical Sciences, University of Hyderabad, Hyderabad, India
| | - Rachid El Fatimy
- Department of Neurology, Center for Neurologic Diseases, Initiative for RNA Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Ionita Ghiran
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Boston, Mass
| | | | - Miguel A de la Fuente
- Instituto de Biología y Genética Molecular, University of Valladolid, Valladolid, Spain
| | - Haruhito Azuma
- Department of Urology, Osaka Medical College, Osaka, Japan
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Anju Vasudevan
- Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital, Harvard Medical School, Belmont, Mass
| | - Abdallah Elkhal
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
50
|
Carisey AF, Mace EM, Saeed MB, Davis DM, Orange JS. Nanoscale Dynamism of Actin Enables Secretory Function in Cytolytic Cells. Curr Biol 2018; 28:489-502.e9. [PMID: 29398219 PMCID: PMC5835143 DOI: 10.1016/j.cub.2017.12.044] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/13/2017] [Accepted: 12/20/2017] [Indexed: 11/21/2022]
Abstract
Natural killer (NK) cells are innate immune effectors that lyse virally infected and tumorigenic cells through the formation of an immunological synapse. Actin remodeling at the lytic immunological synapse is a critical requirement for multiple facets of cytotoxic function. Activating receptor and integrin signaling leads to the regulated turnover and remodeling of actin, which is required for adhesion, sustained receptor signaling, and ultimately exocytosis. NK cells undergo lytic granule exocytosis in hypodense regions of a pervasive actin network. Although these requirements have been well demonstrated, neither the dynamic regulation of synaptic actin nor its specific function, however, has been determined at a nanoscale level. Here, live-cell super-resolution microscopy demonstrates nanoscale filamentous actin dynamism in NK cell lytic granule secretion. Following cell spreading, the overall content of the branched actin network at an immune synapse is stable over time and contains branched actin fibers and discrete actin foci. Similar actin architecture is generated in cytolytic T cells, although the timescale differs from that of NK cells. Individual filament displacement leads to stochastic clearance formation and disappearance, which are independent of lytic granule positioning. Actin dynamism is dependent upon branched network formation mediated by Arp2/3 and contractility generated by myosin IIA. Importantly, the use of small-molecule inhibitors demonstrates that actin dynamism is ultimately needed for granule secretion. Thus, we describe a requirement for nanoscale actin fiber rearrangement in generating the complex actin architecture that enables lytic granule secretion.
Collapse
Affiliation(s)
- Alexandre F Carisey
- Center for Human Immunobiology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX 77030, USA; Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Emily M Mace
- Center for Human Immunobiology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX 77030, USA
| | - Mezida B Saeed
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Daniel M Davis
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Jordan S Orange
- Center for Human Immunobiology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|