162 results on '"Arian, Laurence"'
Search Results
2. Pembrolizumab for the treatment of progressive multifocal leukoencephalopathy following anti‐CD19 CAR‐T therapy: a case report
- Author
-
Strachan Mackenzie, Manar Shafat, Harriet Roddy, Harpreet Hyare, Lorna Neill, Maria A. V. Marzolini, Michael Gilhooley, Teresa Marafioti, Eleanna Kara, Emilie Sanchez, Jeremy Rees, David S. Lynch, Kirsty Thomson, Kirit M. Ardeshna, Arian Laurence, Karl S. Peggs, Maeve O'Reilly, and Claire Roddie
- Subjects
checkpoint inhibition ,chimeric antigen receptor T‐ cell ,herald lesion ,immune reconstitution inflammatory syndrome ,pembrolizumab ,progressive multifocal leukoencephalopathy ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Progressive multifocal leukoencephalopathy (PML) is an opportunistic brain infection with few treatment options and poor survival when reversal of the underlying immune dysfunction is not achievable. JC polyomavirus reactivation resulting in PML can rarely complicate chimeric antigen receptor T‐cell (CAR‐T) therapy. We describe successful treatment of PML with Programmed death‐1 (PD‐1) blockade using pembrolizumab, 4 months following axicabtagene ciloleucel. Radiological features of immune reconstitution inflammatory syndrome without clinical deterioration were seen. Evidence of anti‐viral immune reconstitution by in vitro detection of JC‐specific T‐cells and sustained neurological recovery in this patient suggest PD‐1 blockade may be an effective treatment approach for PML post‐CAR‐T.
- Published
- 2021
- Full Text
- View/download PDF
3. Viral integration drives multifocal HCC during the occult HBV infection
- Author
-
Xiao-Ping Chen, Xin Long, Wen-long Jia, Han-Jie Wu, Jing Zhao, Hui-Fang Liang, Arian Laurence, Jun Zhu, Dong Dong, Yan Chen, Long Lin, Yu-Dong Xia, Wei-Yang Li, Gui-Bo Li, Zhi-Kun Zhao, Kui Wu, Yong Hou, Jing-Jing Yu, Wei Xiao, Guo-Ping Wang, Peng-Cheng Zhu, Wei Chen, Ming-Zhou Bai, Yi-Xing Jian, Karsten Kristiansen, and Qian Chen
- Subjects
Hepatitis B ,Hepatocellular carcinoma ,Single-cell sequencing ,Viral integration ,Virome capture sequencing ,Whole genome sequencing ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background & Aims Although the prognosis of patients with occult hepatitis B virus (HBV) infection (OBI) is usually benign, a small portion may undergo cirrhosis and subsequently hepatocellular carcinoma (HCC). We studied the mechanism of life-long Integration of virus DNA into OBI host’s genome, of which may induce hepatocyte transformation. Methods We applied HBV capture sequencing on single cells from an OBI patient who, developed multiple HCC tumors and underwent liver resection in May 2013 at Tongji Hospital in China. Despite with the undetectable virus DNA in serum, we determined the pattern of viral integration in tumor cells and adjacent non-tumor cells and obtained the details of the viral arrangement in host genome, and furthermore the HBV integrated region in cancer genome. Results HBV captured sequencing of tissues and individual cells revealed that samples from multiple tumors shared two viral integration sites that could affect three host genes, including CSMD2 on chr1 and MED30/EXT1 on chr8. Whole genome sequencing further indicated one hybrid chromosome formed by HBV integrations between chr1 and chr8 that was shared by multiple tumors. Additional 50 poorly differentiated liver tumors and the paired adjacent non-tumors were evaluated and functional studies suggested up-regulated EXT1 expression promoted HCC growth. We further observed that the most somatic mutations within the tumor cell genome were common among the multiple tumors, suggesting that HBV associated, multifocal HCC is monoclonal in origin. Conclusion Through analyzing the HBV integration sites in multifocal HCC, our data suggested that the tumor cells were monoclonal in origin and formed in the absence of active viral replication, whereas the affected host genes may subsequently contribute to carcinogenesis.
- Published
- 2019
- Full Text
- View/download PDF
4. ATP6V0d2 mediates leucine-induced mTORC1 activation and polarization of macrophages
- Author
-
Pingfei Li, Xiaofei Deng, Jing Luo, Yufei Chen, Guoyu Bi, Feili Gong, Zhengping Wei, Na Liu, Huabin Li, Arian Laurence, and Xiang-Ping Yang
- Subjects
Cytology ,QH573-671 ,Animal biochemistry ,QP501-801 - Published
- 2019
- Full Text
- View/download PDF
5. Protein kinases: drug targets for immunological disorders
- Author
-
Leslie Castelo-Soccio, Hanna Kim, Massimo Gadina, Pamela L. Schwartzberg, Arian Laurence, and John J. O’Shea
- Subjects
History ,Computer Science Applications ,Education - Published
- 2023
- Full Text
- View/download PDF
6. OA22 Management of haemophagocytic lymphohistiocytosis: service evaluation of a national multi-disciplinary meeting
- Author
-
Neal S Shah, Matthew Hutchinson, Samuel Clark, Emilie Sanchez, Michael Brown, Neil Stone, Aisling S Carr, Ben Carpenter, Arian Laurence, Satyen H Gohil, Rachel S Tattersall, Jessica J Manson, and Alexis Jones
- Subjects
Rheumatology ,Pharmacology (medical) - Abstract
Background/Aims Haemophagocytic lymphohistiocytosis (HLH) is a devastating condition caused by uncontrolled activation of the immune system. If left untreated, the condition leads to multi-organ failure and death. Even with treatment, recent UK data has shown a mortality rate of 50%. In 2019, a group of clinicians from University College London Hospital (UCLH) came together with the aim of improving outcomes for patients with HLH. The UCLH HLH multi-disciplinary meeting (MDM) has been running since that time and includes representation from rheumatology, haematology, infectious diseases, tropical medicine, virology, neurology, and critical care. Clinicians from any UK trust are invited to join and present patients. As part of a service evaluation project, we aimed to assess the scope of this meeting with regards to the number, demographics and primary diagnoses of patients discussed and the geographical location of the referring clinical team. Methods This is a retrospective descriptive analysis of all patients discussed at the UCLH HLH MDT from 2nd September 2020 to 20th July 2022. Data were obtained from electronic health records and analysed in Prism version 9.4.1. This work has been registered as a service evaluation project within the Division of Medical Specialities at UCLH. Results 93 patients were discussed. 38 were female. 55 were male. The median age was 38 years. Triggers for HLH were haematological malignancy (38.0%), infection (29.3%) rheumatological (14.1%) primary HLH (6.5%) and unknown (7.6%). In 4 patients, HLH was not felt to be the underlying diagnosis. 40 patients were managed as inpatients at UCLH, 37 of whom were transferred from other trusts. Geographical data were obtained on 81 patients. Patients were referred from all 9 regions of England: London (34), North East (1), North West (2), Yorkshire (2), East Midlands (1), West Midlands (3) South East (11), East of England (11), South West (11) as well as Wales (2), Scotland (2) and Ireland (1). Haemophagocytosis was confirmed on bone marrow biopsy in 66.7% of patients. Mortality data showed that 32 (34%) patients died following their diagnosis. Conclusion A multi-disciplinary approach is essential in the management of HLH. Our data show that patients with HLH are young, have a high mortality and broad range of pathology. The UCLH HLH MDT currently serves a wide geographical area across the UK and UCLH acts as a tertiary referral centre for patients with the condition. Disclosure N.S. Shah: None. M. Hutchinson: None. S. Clark: None. E. Sanchez: None. M. Brown: None. N. Stone: None. A.S. Carr: None. B. Carpenter: None. A. Laurence: None. S.H. Gohil: None. R.S. Tattersall: None. J.J. Manson: None. A. Jones: None.
- Published
- 2023
- Full Text
- View/download PDF
7. P004 Infective complications in patients with secondary haemophagocytic lymphohistiocytosis: a case series from a large central London teaching hospital
- Author
-
Pratyasha Saha, Naina McCann, Michael Brown, Neil Stone, Emilie Sanchez, Ben Carpenter, Arian Laurence, Matthew Hutchinson, Alexis Jones, Satyen H Gohil, and Jessica Manson
- Subjects
Rheumatology ,Pharmacology (medical) - Abstract
Background/Aims Haemophagocytic lymphohistiocytosis (HLH) is a rare systemic inflammatory condition that is characterised by excessive macrophage activation in response to a wide range of triggers, including infections, rheumatological and haematological conditions. HLH is associated with high morbidity and mortality, and treatment requires significant immunosuppression. University College London Hospitals (UCLH) operates an HLH service; we were keen to understand the burden of infective complications in our cohort of patients and establish specific guidelines. Methods A retrospective descriptive analysis was performed of all adults with secondary HLH over a two-year time-frame (1stApril 2019 to 1st April 2021) at UCLH. UCLH has a designated HLH multi-disciplinary team (MDT) supported by rheumatology, infectious diseases, tropical medicine, virology, haematology and critical care. This work was registered as a service evaluation project by the Medical Specialities Division. Data was collated from electronic health records into REDCap, and processed in R version 1.4.1103 and Microsoft Excel by two independent authors. Results 40 patients were identified, with a median age of 37. 14 patients had an identified haematological trigger (35%), 12 an infectious one (30%) and 5 a rheumatological (12.5%). 24 patients (60%) were admitted to intensive care and 13 died during admission (32.5%). 35 patients (87.5%) received steroids at some time during their admission, with 24 (60%) receiving a long course. Four patients (10%) had prolonged neutropaenia during admission. Twenty-four patients (60%) developed an infection during their admission; with 33 bacterial, 7 viral and 12 fungal infections (5 proven, 2 probable and 5 possible) identified. Sixteen patients had >1 infection. Those who did not have a diagnosed infection during their admission were more likely to survive than those who did (93% vs 44%, p = 0.047). The mortality rate of those with fungal infections was 89%, compared to 43% for bacterial and viral infections. Of the 24 patients who received a long course of steroids, 14 (58%) did not receive anti-fungal prophylaxis during their admission and three of these patients were diagnosed with fungal infections during their admission. Of the 4 patients with prolonged neutropenia, 1 (25%) did not receive antifungal prophylaxis and did suffer from a confirmed fungal infection. One patient with confirmed PCP had not previously been taking PCP prophylaxis. Of the seven patients with viral infections, 2 had HSV-1 infections and neither had received anti-viral prophylaxis before their infection start date. Conclusion We demonstrate that a high proportion of patients with HLH develop infective complications, which is associated with increased mortality. Prior to this analysis, the use of antimicrobial prophylaxis was variable. We have since written a specific guideline for the routine use of antimicrobial prophylaxis for patients with HLH, with a view to auditing future care against this guideline. Disclosure P. Saha: None. N. McCann: None. M. Brown: None. N. Stone: None. E. Sanchez: None. B. Carpenter: None. A. Laurence: None. M. Hutchinson: None. A. Jones: None. S.H. Gohil: None. J. Manson: None.
- Published
- 2023
- Full Text
- View/download PDF
8. Figure S2 from TFEB Mediates Immune Evasion and Resistance to mTOR Inhibition of Renal Cell Carcinoma via Induction of PD-L1
- Author
-
Xiang-Ping Yang, Bing Li, Arian Laurence, Fang Zheng, Guoping Wang, Jie Wu, Xiang Cheng, Zhaohui Tang, Chaoyang Sun, Jing Wang, Na Liu, Zhengping Wei, Qian Zhang, Shuaishuai Chai, Lu Zheng, Yufei Chen, Yuting Dong, Minghui Xia, Yaqi Duan, and Cai Zhang
- Abstract
No effect of TFEB on RCC tumor growth in nude mice
- Published
- 2023
- Full Text
- View/download PDF
9. Data from TFEB Mediates Immune Evasion and Resistance to mTOR Inhibition of Renal Cell Carcinoma via Induction of PD-L1
- Author
-
Xiang-Ping Yang, Bing Li, Arian Laurence, Fang Zheng, Guoping Wang, Jie Wu, Xiang Cheng, Zhaohui Tang, Chaoyang Sun, Jing Wang, Na Liu, Zhengping Wei, Qian Zhang, Shuaishuai Chai, Lu Zheng, Yufei Chen, Yuting Dong, Minghui Xia, Yaqi Duan, and Cai Zhang
- Abstract
Purpose:Despite the FDA approval of mTOR inhibitors (mTORi) for the treatment of renal cell carcinoma (RCC), the benefits are relatively modest and the few responders usually develop resistance. We investigated whether the resistance to mTORi is due to upregulation of PD-L1 and the underlying molecular mechanism.Experimental Design:The effects of transcription factor EB (TFEB) on RCC proliferation, apoptosis, and migration were evaluated. Correlation of TFEB with PD-L1 expression, as well as effects of mTOR inhibition on TFEB and PD-L1 expression, was assessed in human primary clear cell RCCs. The regulation of TFEB on PD-L1 was assessed by chromatin immunoprecipitation and luciferase reporter assay. The therapeutic efficacies of mTORi plus PD-L1 blockade were evaluated in a mouse model. The function of tumor-infiltrating CD8+ T cells was analyzed by flow cytometry.Results:TFEB did not affect tumor cell proliferation, apoptosis, and migration. We found a positive correlation between TFEB and PD-L1 expression in RCC tumor tissues, primary tumor cells, and RCC cells. TFEB bound to PD-L1 promoter in RCCs and inhibition of mTOR led to enhanced TFEB nuclear translocation and PD-L1 expression. Simultaneous inhibition of mTOR and blockade of PD-L1 enhanced CD8+ cytolytic function and tumor suppression in a xenografted mouse model of RCC.Conclusions:These data revealed that TFEB mediates resistance to mTOR inhibition via induction of PD-L1 in human primary RCC tumors, RCC cells, and murine xenograft model. Our data provide a strong rationale to target mTOR and PD-L1 jointly as a novel immunotherapeutic approach for RCC treatment.
- Published
- 2023
- Full Text
- View/download PDF
10. Table S1 from TFEB Mediates Immune Evasion and Resistance to mTOR Inhibition of Renal Cell Carcinoma via Induction of PD-L1
- Author
-
Xiang-Ping Yang, Bing Li, Arian Laurence, Fang Zheng, Guoping Wang, Jie Wu, Xiang Cheng, Zhaohui Tang, Chaoyang Sun, Jing Wang, Na Liu, Zhengping Wei, Qian Zhang, Shuaishuai Chai, Lu Zheng, Yufei Chen, Yuting Dong, Minghui Xia, Yaqi Duan, and Cai Zhang
- Abstract
Patient characteristics
- Published
- 2023
- Full Text
- View/download PDF
11. Data from Transforming Growth Factor β Subverts the Immune System into Directly Promoting Tumor Growth through Interleukin-17
- Author
-
Lalage M. Wakefield, Jay A. Berzofsky, Scott Lonning, Mizuko Mamura, Aleksandra Michalowska, Arian Laurence, Yu-an Yang, Nga Voong, Helen Chae, Mi-Jin Kang, Masaki Terabe, and Jeong-Seok Nam
- Abstract
Overexpression of the immunosuppressive cytokine transforming growth factor β (TGF-β) is one strategy that tumors have developed to evade effective immunesurveillance. Using transplantable models of breast and colon cancer, we made the unexpected finding that CD8+ cells in tumor-bearing animals can directly promote tumorigenesis, by a mechanism that is dependent on TGF-β. We showed that CD8+ splenocytes from tumor-bearing mice expressed elevated interleukin (IL)-17 when compared with naive mice, and that CD8+ T cells could be induced to make IL-17 on addition of TGF-β and IL-6 in vitro. Treatment of mice with anti–TGF-β antibodies in vivo reduced IL-17 expression both in the tumor and the locoregional lymph nodes. Although IL-17 has not previously been shown to act as a survival factor for epithelial cells, we found that IL-17 suppressed apoptosis of several tumor cell lines in vitro, suggesting that this altered T-cell polarization has the potential to promote tumorigenesis directly, rather than indirectly through inflammatory sequelae. Consistent with this hypothesis, knockdown of the IL-17 receptor in 4T1 mouse mammary cancer cells enhanced apoptosis and decreased tumor growth in vivo. Thus, in addition to suppressing immune surveillance, tumor-induced TGF-β may actively subvert the CD8+ arm of the immune system into directly promoting tumor growth by an IL-17–dependent mechanism. [Cancer Res 2008;68(10):3915–23]
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figures 1-5, Table 1 from Transforming Growth Factor β Subverts the Immune System into Directly Promoting Tumor Growth through Interleukin-17
- Author
-
Lalage M. Wakefield, Jay A. Berzofsky, Scott Lonning, Mizuko Mamura, Aleksandra Michalowska, Arian Laurence, Yu-an Yang, Nga Voong, Helen Chae, Mi-Jin Kang, Masaki Terabe, and Jeong-Seok Nam
- Abstract
Supplementary Figures 1-5, Table 1 from Transforming Growth Factor β Subverts the Immune System into Directly Promoting Tumor Growth through Interleukin-17
- Published
- 2023
- Full Text
- View/download PDF
13. Genomic diagnosis and care co-ordination for monogenic inflammatory bowel disease in children and adults: Consensus guideline on behalf of the British Society of Gastroenterology and British Society of Paediatric Gastroenterology, Hepatology and Nutrition
- Author
-
Jochen Kammermeier, Christopher A Lamb, Kelsey D J Jones, Carl A Anderson, Emma L Baple, Chrissy Bolton, Helen Braggins, Tanya I Coulter, Kimberly C Gilmour, Vicki Gregory, Sophie Hambleton, David Hartley, A Barney Hawthorne, Sarah Hearn, Arian Laurence, Miles Parkes, Richard K Russell, R Alexander Speight, Simon Travis, David C Wilson, and Holm H Uhlig
- Subjects
Hepatology ,Gastroenterology - Abstract
Genomic medicine enables the identification of patients with rare or ultra-rare monogenic forms of inflammatory bowel disease (IBD) and supports clinical decision making. Patients with monogenic IBD frequently experience extremely early onset of treatment-refractory disease, with complex extraintestinal disease typical of immunodeficiency. Since more than 100 monogenic disorders can present with IBD, new genetic disorders and variants are being discovered every year, and as phenotypic expression of the gene defects is variable, adaptive genomic technologies are required. Monogenic IBD has become a key area to establish the concept of precision medicine. Clear guidance and standardised, affordable applications of genomic technologies are needed to implement exome or genome sequencing in clinical practice. This joint British Society of Gastroenterology and British Society of Paediatric Gastroenterology, Hepatology and Nutrition guideline aims to ensure that testing resources are appropriately applied to maximise the benefit to patients on a national scale, minimise health-care disparities in accessing genomic technologies, and optimise resource use. We set out the structural requirements for genomic medicine as part of a multidisciplinary team approach. Initiation of genomic diagnostics should be guided by diagnostic criteria for the individual patient, in particular the age of IBD onset and the patient's history, and potential implications for future therapies. We outline the diagnostic care pathway for paediatric and adult patients. This guideline considers how to handle clinically actionable findings in research studies and the impact of consumer-based genomics for monogenic IBD. This document was developed by multiple stakeholders, including UK paediatric and adult gastroenterology physicians, immunologists, transplant specialists, clinical geneticists, scientists, and research leads of UK genetic programmes, in partnership with patient representatives of several IBD and rare disease charities.
- Published
- 2023
- Full Text
- View/download PDF
14. Correction to: Viral integration drives multifocal HCC during the occult HBV infection
- Author
-
Xiao-Ping Chen, Xin Long, Wen-long Jia, Han-Jie Wu, Jing Zhao, Hui-Fang Liang, Arian Laurence, Jun Zhu, Dong Dong, Yan Chen, Long Lin, Yu-Dong Xia, Wei-Yang Li, Gui-Bo Li, Zhi-Kun Zhao, Kui Wu, Yong Hou, Jing-Jing Yu, Wei Xiao, Guo-Ping Wang, Peng-Cheng Zhu, Wei Chen, Ming-Zhou Bai, Yi-Xing Jian, Karsten Kristiansen, and Qian Chen
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
In the original publication of this article [1], Fig. 6 is wrong and the updated figure is shown below.
- Published
- 2019
- Full Text
- View/download PDF
15. Transcription factor EB coordinates environmental cues to regulate T regulatory cells' mitochondrial fitness and function
- Author
-
Minghui Xia, Cai Zhang, Yufei Chen, Xiujuan Zhao, Si Zhang, Yang Liu, You Cai, Zhengping Wei, Qiuyang Du, Wenting Yu, Chun Zhou, Hongjun Xiao, Guihua Wang, Xiang Cheng, Heng Mei, Arian Laurence, Jing Wang, Yu Hu, Huabin Li, and Xiang-Ping Yang
- Subjects
Disease Models, Animal ,Mice ,Multidisciplinary ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Neoplasms ,Autophagy ,Animals ,Interleukin-2 ,Adaptation, Physiological ,T-Lymphocytes, Regulatory ,Xenograft Model Antitumor Assays ,Autoimmune Diseases ,Mitochondria - Abstract
T regulatory (Treg) cells are essential for self-tolerance whereas they are detrimental for dampening the host anti-tumor immunity. How Treg cells adapt to environmental signals to orchestrate their homeostasis and functions remains poorly understood. Here, we identified that transcription factor EB (TFEB) is induced by host nutrition deprivation or interleukin (IL)-2 in CD4 + T cells. The loss of TFEB in Treg cells leads to reduced Treg accumulation and impaired Treg function in mouse models of cancer and autoimmune disease. TFEB intrinsically regulates genes involved in Treg cell differentiation and mitochondria function while it suppresses expression of proinflammatory cytokines independently of its established roles in autophagy. This coordinated action is required for mitochondria integrity and appropriate lipid metabolism in Treg cells. These findings identify TFEB as a critical regulator for orchestrating Treg generation and function, which may contribute to the adaptive responses of T cells to local environmental cues.
- Published
- 2023
16. Protein Kinase Antagonists
- Author
-
Arian Laurence, Massimo Gadina, Pamela L. Schwartzberg, and John J. O'Shea
- Published
- 2023
- Full Text
- View/download PDF
17. Autocrine Vitamin D-signaling switches off pro-inflammatory programs of Th1 cells
- Author
-
Alexandra F. Freeman, Majid Kazemian, Jack A. Bibby, Daniel Chauss, Daniel S. Chertow, Michail S. Lionakis, Reuben McGregor, Tilo Freiwald, Nehal N. Mehta, Heather L. Teague, Luopin Wang, Audrey Kelly, Behdad Afzali, Estefania Nova-Lamperti, Kevin M. Vannella, Amna Malik, Daniella M. Schwartz, Bingyu Yan, Claudia Kemper, Didier Portilla, Giovanna Lombardi, Marcos J Ramos-Benitez, Susan D. John, Nichola Cooper, Arian Laurence, Paul Lavender, Erin E. West, Zonghao Zhang, and Dhaneshwar Kumar
- Subjects
T-Lymphocytes ,Immunology ,Cell ,Complement ,BACH2 ,Article ,STAT3 ,03 medical and health sciences ,0302 clinical medicine ,Vitamin D and neurology ,medicine ,Immunology and Allergy ,Humans ,Epigenetics ,Vitamin D ,Receptor ,Autocrine signalling ,Transcription factor ,030304 developmental biology ,Inflammation ,0303 health sciences ,biology ,Chemistry ,c-JUN ,COVID-19 ,Cell biology ,single cell RNA-sequencing ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,SARS-CoV2 ,biology.protein ,Homeostasis - Abstract
The molecular mechanisms governing orderly shutdown and retraction of CD4+ type 1 helper T (TH1) cell responses remain poorly understood. Here we show that complement triggers contraction of TH1 responses by inducing intrinsic expression of the vitamin D (VitD) receptor and the VitD-activating enzyme CYP27B1, permitting T cells to both activate and respond to VitD. VitD then initiated the transition from pro-inflammatory interferon-γ+ TH1 cells to suppressive interleukin-10+ cells. This process was primed by dynamic changes in the epigenetic landscape of CD4+ T cells, generating super-enhancers and recruiting several transcription factors, notably c-JUN, STAT3 and BACH2, which together with VitD receptor shaped the transcriptional response to VitD. Accordingly, VitD did not induce interleukin-10 expression in cells with dysfunctional BACH2 or STAT3. Bronchoalveolar lavage fluid CD4+ T cells of patients with COVID-19 were TH1-skewed and showed de-repression of genes downregulated by VitD, from either lack of substrate (VitD deficiency) and/or abnormal regulation of this system. During homeostasis TH1 cells activate a cell-intrinsic inflammatory shutdown program and shift to IL-10 production. Chauss et al. find that this TH1 homeostatic program is dependent on vitamin D signaling and is disrupted in severe COVID-19.
- Published
- 2021
18. Tbet is a critical modulator of FoxP3 expression in autoimmune graft-versus-host disease
- Author
-
Shoba Amarnath, Arian Laurence, Nathaniel Zhu, Renato Cunha, Michael A. Eckhaus, Samuel Taylor, Jason E. Foley, Monalisa Ghosh, Tania C. Felizardo, and Daniel H. Fowler
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
CD4+ T-helper subsets drive autoimmune chronic graft-versus-host disease, a major complication after allogeneic bone marrow transplantation. However, it remains unclear how specific T-helper subsets contribute to chronic graft-versus-host disease. T-helper type 1 cells are one of the major disease-mediating T-cell subsets and require interferon-γ signaling and Tbet expression for their function. Regulatory T cells on the other hand can inhibit T-helper type 1 cell-mediated responses. Using an established murine model that isolates the autoimmune component of graft-versus-host disease, we hypothesized that T-helper type 1 cells would restrict FoxP3-driven regulatory T cells. Upon transfer into immune-deficient syngeneic hosts, alloreactive Tbx21−/−CD4+ T cells led to marked increases in FoxP3+ cells and reduced clinical evidence of autoimmunity. To evaluate whether peripheral induction contributed to regulatory T-cell predominance, we adoptively transferred Tbx21−/− T cells that consisted of fate mapping for FoxP3: recipients of flow-purified effector cells that were Foxp3− and Tbx21−/− had enhanced T-regulatory-cell predominance during autoimmune graft-versus-host disease. These data directly demonstrated that peripheral T-regulatory-cell induction was inhibited by Tbet. Finally, Tbx21−/− T-regulatory cells cross-regulated autoimmune wild-type T-effector-cell cytokine production in vivo. The Tbet pathway therefore directly impairs T-regulatory-cell reconstitution and is consequently a feasible target in efforts to prevent autoimmune graft-versus-host disease.
- Published
- 2017
- Full Text
- View/download PDF
19. Allogeneic stem cell transplantation compared to conservative management in adults with inborn errors of immunity
- Author
-
Morgane Cheminant, Thomas A. Fox, Mickael Alligon, Olivier Bouaziz, Bénédicte Neven, Despina Moshous, Stéphane Blanche, Aurélien Guffroy, Claire Fieschi, Marion Malphettes, Nicolas Schleinitz, Antoinette Perlat, Jean-François Viallard, Nathalie Dhedin, Françoise Sarrot-Reynauld, Isabelle Durieu, Sébastien Humbert, Fanny Fouyssac, Vincent Barlogis, Benjamin Carpenter, Rachael Hough, Arian Laurence, Ambroise Marçais, Ronjon Chakraverty, Olivier Hermine, Alain Fischer, Siobhan O. Burns, Nizar Mahlaoui, Emma C. Morris, and Felipe Suarez
- Subjects
Adult ,Transplantation Conditioning ,Immunology ,Hematopoietic Stem Cell Transplantation ,Graft vs Host Disease ,Cell Biology ,Hematology ,Granulomatous Disease, Chronic ,Conservative Treatment ,Biochemistry ,Young Adult ,Humans ,Transplantation, Homologous ,Retrospective Studies ,Stem Cell Transplantation - Abstract
Allogeneic hematopoietic stem cell transplantation (alloSCT) is curative for severe inborn errors of immunity (IEIs), with recent data suggesting alloSCT in adulthood is safe and effective in selected patients. However, questions remain regarding the indications for and optimal timing of transplant. We retrospectively compared outcomes of transplanted vs matched nontransplanted adults with severe IEIs. Seventy-nine patients (aged ≥ 15 years) underwent alloSCT between 2008 and 2018 for IEIs such as chronic granulomatous disease (n = 20) and various combined immune deficiencies (n = 59). A cohort of nontransplanted patients from the French Centre de Référence Déficits Immunitaires Héréditaires registry was identified blindly for case-control analysis, with ≤3 matched controls per index patient, without replacement. The nontransplanted patients were matched for birth decade, age at last review greater than index patient age at alloSCT, chronic granulomatous disease or combined immune deficiencies, and autoimmune/lymphoproliferative complications. A total of 281 patients were included (79 transplanted, 202 nontransplanted). Median age at transplant was 21 years. Transplant indications were mainly lymphoproliferative disease (n = 23) or colitis (n = 15). Median follow-up was 4.8 years (interquartile range, 2.5-7.2). One-year transplant-related mortality rate was 13%. Estimated disease-free survival at 5 years was higher in transplanted patients (58% vs 33%; P = .007). Nontransplanted patients had an ongoing risk of severe events, with an increased mean cumulative number of recurrent events compared with transplanted patients. Sensitivity analyses removing patients with common variable immune deficiency and their matched transplanted patients confirm these results. AlloSCT prevents progressive morbidity associated with IEIs in adults, which may outweigh the negative impact of transplant-related mortality.
- Published
- 2022
20. Signal transducer and activator of transcription 5 (STAT5) paralog dose governs T cell effector and regulatory functions
- Author
-
Alejandro Villarino, Arian Laurence, Gertraud W Robinson, Michael Bonelli, Barbara Dema, Behdad Afzali, Han-Yu Shih, Hong-Wei Sun, Stephen R Brooks, Lothar Hennighausen, Yuka Kanno, and John J O'Shea
- Subjects
T cells ,autoimmunity ,STAT5 ,cytokine ,STAT signaling ,transcription factors ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The transcription factor STAT5 is fundamental to the mammalian immune system. However, the relationship between its two paralogs, STAT5A and STAT5B, and the extent to which they are functionally distinct, remain uncertain. Using mouse models of paralog deficiency, we demonstrate that they are not equivalent for CD4+ 'helper' T cells, the principal orchestrators of adaptive immunity. Instead, we find that STAT5B is dominant for both effector and regulatory (Treg) responses and, therefore, uniquely necessary for immunological tolerance. Comparative analysis of genomic distribution and transcriptomic output confirm that STAT5B has fargreater impact but, surprisingly, the data point towards asymmetric expression (i.e. paralog dose), rather than distinct functional properties, as the key distinguishing feature. Thus, we propose a quantitative model of STAT5 paralog activity whereby relative abundance imposes functional specificity (or dominance) in the face of widespread structural homology.
- Published
- 2016
- Full Text
- View/download PDF
21. Inhibition of acylglycerol kinase sensitizes DLBCL to venetoclax via upregulation of FOXO1-mediated BCL-2 expression
- Author
-
Na Ning, Si Zhang, Qi Wu, Xun Li, Dong Kuang, Yaqi Duan, Minghui Xia, Huicheng Liu, Junmei Weng, Hongping Ba, Zhaohui Tang, Xiang Cheng, Heng Mei, Liu Huang, Qilin Ao, Guoping Wang, Yu Hu, Arian Laurence, Jing Wang, Guihua Wang, and Xiang-Ping Yang
- Subjects
Sulfonamides ,Forkhead Box Protein O1 ,Medicine (miscellaneous) ,Apoptosis ,Bridged Bicyclo Compounds, Heterocyclic ,Up-Regulation ,Mice ,Phosphotransferases (Alcohol Group Acceptor) ,Proto-Oncogene Proteins c-bcl-2 ,Cell Line, Tumor ,Animals ,Humans ,Lymphoma, Large B-Cell, Diffuse ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) - Published
- 2022
22. Retrospective, Landmark Analysis of Long-term Adult Morbidity Following Allogeneic HSCT for Inborn Errors of Immunity in Infancy and Childhood
- Author
-
James W. Day, Reem Elfeky, Bethany Nicholson, Rupert Goodman, Rachel Pearce, Thomas A. Fox, Austen Worth, Claire Booth, Paul Veys, Ben Carpenter, Rachael Hough, H. Bobby Gaspar, Penny Titman, Deborah Ridout, Sarita Workman, Fernando Hernandes, Kit Sandford, Arian Laurence, Mari Campbell, Siobhan O. Burns, and Emma C. Morris
- Subjects
Adult ,Transplantation Conditioning ,Immunology ,Hematopoietic Stem Cell Transplantation ,Immunology and Allergy ,Graft vs Host Disease ,Humans ,Morbidity ,Chimerism ,Retrospective Studies - Abstract
Abstract Purpose Allogeneic hematopoietic stem cell transplant (HSCT) remains the treatment of choice for patients with inborn errors of immunity (IEI). There is little published medical outcome data assessing late medical complications following transition to adult care. We sought to document event-free survival (EFS) in transplanted IEI patients reaching adulthood and describe common late-onset medical complications and factors influencing EFS. Methods In this landmark analysis, 83 adults surviving 5 years or more following prior HSCT in childhood for IEI were recruited. The primary endpoint was event-free survival, defined as time post-first HSCT to graft failure, graft rejection, chronic infection, life-threatening or recurrent infections, malignancy, significant autoimmune disease, moderate to severe GVHD or major organ dysfunction. All events occurring less than 5 years post-HSCT were excluded. Results EFS was 51% for the whole cohort at a median of 20 years post HSCT. Multivariable analysis identified age at transplant and whole blood chimerism as independent predictors of long-term EFS. Year of HSCT, donor, conditioning intensity and underlying diagnosis had no significant impact on EFS. 59 events occurring beyond 5 years post-HSCT were documented in 37 patients (45% cohort). A total of 25 patients (30% cohort) experienced ongoing significant complications requiring active medical intervention at last follow-up. Conclusion Although most patients achieved excellent, durable immune reconstitution with infrequent transplant-related complications, very late complications are common and associated with mixed chimerism post-HSCT. Early intervention to correct mixed chimerism may improve long-term outcomes and adult health following HSCT for IEI in childhood.
- Published
- 2021
23. TFEB Mediates Immune Evasion and Resistance to mTOR Inhibition of Renal Cell Carcinoma via Induction of PD-L1
- Author
-
Fang Zheng, Na Liu, Guo-Ping Wang, Chaoyang Sun, Arian Laurence, Yaqi Duan, Xiang Cheng, Qian Zhang, Lu Zheng, Bing Li, Yuting Dong, Shuaishuai Chai, Zhaohui Tang, Jie Wu, Xiang-Ping Yang, Minghui Xia, Cai Zhang, Zhengping Wei, Jing Wang, and Yufei Chen
- Subjects
0301 basic medicine ,Cancer Research ,Gene Expression ,Apoptosis ,urologic and male genital diseases ,B7-H1 Antigen ,Flow cytometry ,Mice ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Downregulation and upregulation ,Cell Movement ,Cell Line, Tumor ,PD-L1 ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Carcinoma, Renal Cell ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,medicine.diagnostic_test ,biology ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Chemistry ,TOR Serine-Threonine Kinases ,Xenograft Model Antitumor Assays ,Kidney Neoplasms ,female genital diseases and pregnancy complications ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Cell culture ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,TFEB ,Tumor Escape ,Chromatin immunoprecipitation - Abstract
Purpose: Despite the FDA approval of mTOR inhibitors (mTORi) for the treatment of renal cell carcinoma (RCC), the benefits are relatively modest and the few responders usually develop resistance. We investigated whether the resistance to mTORi is due to upregulation of PD-L1 and the underlying molecular mechanism. Experimental Design: The effects of transcription factor EB (TFEB) on RCC proliferation, apoptosis, and migration were evaluated. Correlation of TFEB with PD-L1 expression, as well as effects of mTOR inhibition on TFEB and PD-L1 expression, was assessed in human primary clear cell RCCs. The regulation of TFEB on PD-L1 was assessed by chromatin immunoprecipitation and luciferase reporter assay. The therapeutic efficacies of mTORi plus PD-L1 blockade were evaluated in a mouse model. The function of tumor-infiltrating CD8+ T cells was analyzed by flow cytometry. Results: TFEB did not affect tumor cell proliferation, apoptosis, and migration. We found a positive correlation between TFEB and PD-L1 expression in RCC tumor tissues, primary tumor cells, and RCC cells. TFEB bound to PD-L1 promoter in RCCs and inhibition of mTOR led to enhanced TFEB nuclear translocation and PD-L1 expression. Simultaneous inhibition of mTOR and blockade of PD-L1 enhanced CD8+ cytolytic function and tumor suppression in a xenografted mouse model of RCC. Conclusions: These data revealed that TFEB mediates resistance to mTOR inhibition via induction of PD-L1 in human primary RCC tumors, RCC cells, and murine xenograft model. Our data provide a strong rationale to target mTOR and PD-L1 jointly as a novel immunotherapeutic approach for RCC treatment.
- Published
- 2019
- Full Text
- View/download PDF
24. Progressive multifocal leukoencephalopathy in the era of chimeric antigen receptor T-cell therapy
- Author
-
Claire Roddie, Maeve A O'Reilly, Arian Laurence, Strachan Mackenzie, and Karl S. Peggs
- Subjects
Receptors, Chimeric Antigen ,business.industry ,Progressive multifocal leukoencephalopathy ,medicine ,Cancer research ,Cell- and Tissue-Based Therapy ,Leukoencephalopathy, Progressive Multifocal ,Humans ,Chimeric Antigen Receptor T-Cell Therapy ,Hematology ,medicine.disease ,business ,Immunotherapy, Adoptive - Published
- 2021
25. SARS-CoV-2 drives JAK1/2-dependent local complement hyperactivation
- Author
-
Richard Gregory, Luopin Wang, Nazish Malik, Nathalie Niyonzima, Charles J. Zhang, Jason R. Spence, Sonja Ghidelli-Disse, Matthew R. Olson, Marcus Bantscheff, Stefania Pittaluga, Tristan Frum, Majid Kazemian, Konstantinos D. Alysandratos, Jonathan Z. Sexton, Daniel Chauss, Darrell N. Kotton, Michail S. Lionakis, Carmen Mirabelli, Erin E. West, Didier Portilla, Bingyu Yan, Eva-Maria Nichols, Christiane E. Wobus, Tilo Freiwald, Shahram Kordasti, Martin Kolev, Behdad Afzali, Jack A. Bibby, Arian Laurence, and Claudia Kemper
- Subjects
0301 basic medicine ,Myeloid ,Immunology ,General Medicine ,Biology ,Complement factor B ,Virus ,C3-convertase ,Complement system ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Interferon ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Receptor ,medicine.drug - Abstract
Patients with coronavirus disease 2019 (COVID-19) present a wide range of acute clinical manifestations affecting the lungs, liver, kidneys and gut. Angiotensin converting enzyme (ACE) 2, the best-characterized entry receptor for the disease-causing virus SARS-CoV-2, is highly expressed in the aforementioned tissues. However, the pathways that underlie the disease are still poorly understood. Here, we unexpectedly found that the complement system was one of the intracellular pathways most highly induced by SARS-CoV-2 infection in lung epithelial cells. Infection of respiratory epithelial cells with SARS-CoV-2 generated activated complement component C3a and could be blocked by a cell-permeable inhibitor of complement factor B (CFBi), indicating the presence of an inducible cell-intrinsic C3 convertase in respiratory epithelial cells. Within cells of the bronchoalveolar lavage of patients, distinct signatures of complement activation in myeloid, lymphoid and epithelial cells tracked with disease severity. Genes induced by SARS-CoV-2 and the drugs that could normalize these genes both implicated the interferon-JAK1/2-STAT1 signaling system and NF-κB as the main drivers of their expression. Ruxolitinib, a JAK1/2 inhibitor, normalized interferon signature genes and all complement gene transcripts induced by SARS-CoV-2 in lung epithelial cell lines, but did not affect NF-κB-regulated genes. Ruxolitinib, alone or in combination with the antiviral remdesivir, inhibited C3a protein produced by infected cells. Together, we postulate that combination therapy with JAK inhibitors and drugs that normalize NF-κB-signaling could potentially have clinical application for severe COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
26. Tissue inhibitor of metalloproteinase 1 is preferentially expressed in Th1 and Th17 T-helper cell subsets and is a direct STAT target gene.
- Author
-
Adewole Adamson, Kamran Ghoreschi, Matthew Rittler, Qian Chen, Hong-Wei Sun, Golnaz Vahedi, Yuka Kanno, William G Stetler-Stevenson, John J O'Shea, and Arian Laurence
- Subjects
Medicine ,Science - Abstract
CD4(+) T helper (Th) cells differentiate into distinct effector subsets that are critical for host defense, but are also implicated in the pathogenesis of autoimmune disorders. Thelper17 (Th17) cells in particular are emerging as important drivers of multiple diseases including psoriasis, spondyloarthropathy and multiple sclerosis. To gain insight into the function of Th17 cells, we performed transcriptional profiling in hopes of elucidating products not previously recognized as being functionally relevant in these T cells. Herein, we demonstrate that tissue inhibitor of metalloproteinase 1 (TIMP1), a secreted protein with pleiotropic effects on cellular growth, survival and integrity of the extracellular matrix, is preferentially produced by Th17 and Th1 cells. We further show that Th1 and Th17 cell TIMP1 regulation follows separate mechanisms with a requirement for STAT4 in the former and STAT3 in the latter. Finally, we demonstrate that when restricted to T cells, expression of TIMP1 promotes neuropathology in experimental allergic encephalomyelitis.
- Published
- 2013
- Full Text
- View/download PDF
27. Inborn errors of IL-6 family cytokine responses
- Author
-
Holm H. Uhlig, James Ed Thaventhiran, Arian Laurence, Yin-Huai Chen, and Sarah Spencer
- Subjects
0301 basic medicine ,STAT3 Transcription Factor ,Glycosylation ,Genotype ,medicine.medical_treatment ,Immunology ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Cytokine Receptor gp130 ,Immunology and Allergy ,Animals ,Humans ,Genetic Predisposition to Disease ,Interleukin 6 ,STAT3 ,Transcription factor ,Alleles ,Genetic Association Studies ,biology ,Interleukin-6 ,Genetic Variation ,Immune dysregulation ,Glycoprotein 130 ,030104 developmental biology ,Cytokine ,Phenotype ,Gene Expression Regulation ,Organ Specificity ,Gain of Function Mutation ,Multigene Family ,biology.protein ,Signal transduction ,Cytokine receptor ,Biomarkers ,030215 immunology ,Signal Transduction ,Transcription Factors - Abstract
The IL-6 family of cytokines mediates functions in host protective immunity, development of multiple organs, tissue regeneration and metabolism. Inborn errors in cytokines or cytokine receptor units highlight specific roles for IL-6, IL-11, LIF, OSM, and CLC signaling whereas incomplete loss-of-function variants in the common receptor chain GP130 encoded by IL6ST or the transcription factor STAT3, as well as genes that affect either GP130 glycosylation (PGM3) or STAT3 transcriptional control (ZNF341) lead to complex phenotypes including features of hyper-IgE syndrome. Gain-of-function variants in the GP130-STAT3 signaling pathway cause immune dysregulation disorders. Insights into IL-6 family cytokine signaling inform on therapeutic application in immune-mediated disorders and potential side effects such as infection susceptibility.
- Published
- 2020
28. Death‐associated protein kinase 1 (DAPK1) controls CD8 + T cell activation, trafficking, and antitumor activity
- Author
-
Qiuyang Du, Zhaohui Tang, Xiang Cheng, Jing Wang, Yufei Chen, Huicheng Liu, Liu Huang, Youming Lu, Arian Laurence, Zhengping Wei, Minghui Xia, Xiang-Ping Yang, Pingfei Li, Guoyu Bi, Ran He, and Huabin Li
- Subjects
0301 basic medicine ,Chemistry ,T cell ,mTORC1 ,Biochemistry ,Cell biology ,Calcineurin ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Death-Associated Protein Kinase 1 ,Genetics ,medicine ,Cytotoxic T cell ,Molecular Biology ,Protein kinase B ,030217 neurology & neurosurgery ,CD8 ,Biotechnology ,Death domain - Abstract
Appropriate migration of cytotoxic T effector cells into the tumors is crucial for their antitumor function. Despite the controversial role of PI3K-Akt in CD8+ T cell mTORC1 activation, a link between Akt-mTORC1 signaling and CD8+ trafficking has been demonstrated. We have recently discovered that TCR-induced calcineurin activates DAPK1, which interacts with TSC2 via its death domain and phosphorylates TSC2 via its kinase domain to mediate mTORC1 activation in CD8+ T cells. However, whether DAPK1 regulates CD8+ trafficking into tumors remains unclear. Here, using pharmacological inhibitor and genetic approaches, we found that like rapamycin, inhibition of DAPK1 activity led to enhanced expression of the homing receptors CD62L and CCR7. Deletion of either kinase domain or death domain in the T cell compartment reduced the T cell activation and maintained the expression of CD62L and CCR7. DAPK1-DD-deficient mice were more susceptible to tumor growth and deficiency of DAPK1 activity significantly reduced the migratory ability of CD8+ into the tumors. These data revealed a crucial role of DAPK1-mTORC1 in mediating CD8+ trafficking and antitumor function.
- Published
- 2020
- Full Text
- View/download PDF
29. An autocrine Vitamin D-driven Th1 shutdown program can be exploited for COVID-19
- Author
-
Heather L. Teague, Susan D. John, Alexandra F. Freeman, Michail S. Lionakis, Paul Lavender, Audrey Kelly, Claudia Kemper, Daniella M. Schwartz, Bingyu Yan, Amna Malik, Jack A. Bibby, Luopin Wang, Daniel Chauss, Nichola Cooper, Estefania Nova-Lamperti, Zonghao Zhang, Tilo Freiwald, Didier Portilla, Giovanna Lombardi, Erin E. West, Behdad Afzali, Arian Laurence, Nehal N. Mehta, Majid Kazemian, and Reuben McGregor
- Subjects
Cell ,Alfacalcidol ,Biology ,Calcitriol receptor ,Article ,chemistry.chemical_compound ,Immune system ,medicine.anatomical_structure ,chemistry ,Immunology ,Vitamin D and neurology ,medicine ,Epigenetics ,Autocrine signalling ,Receptor - Abstract
Pro-inflammatory immune responses are necessary for effective pathogen clearance, but cause severe tissue damage if not shut down in a timely manner1,2. Excessive complement and IFN-γ-associated responses are known drivers of immunopathogenesis3 and are among the most highly induced immune programs in hyper-inflammatory SARS-CoV2 lung infection4. The molecular mechanisms that govern orderly shutdown and retraction of these responses remain poorly understood. Here, we show that complement triggers contraction of IFN-γ producing CD4+ T helper (Th) 1 cell responses by inducing expression of the vitamin D (VitD) receptor (VDR) and CYP27B1, the enzyme that activates VitD, permitting T cells to both activate and respond to VitD. VitD then initiates the transition from pro-inflammatory IFN-γ+ Th1 cells to suppressive IL-10+ Th1 cells. This process is primed by dynamic changes in the epigenetic landscape of CD4+ T cells, generating superenhancers and recruiting c-JUN and BACH2, a key immunoregulatory transcription factor5–7. Accordingly, cells in psoriatic skin treated with VitD increased BACH2 expression, and BACH2 haplo-insufficient CD4+ T cells were defective in IL-10 production. As proof-of-concept, we show that CD4+ T cells in the bronchoalveolar lavage fluid (BALF) of patients with COVID-19 are Th1-skewed and that VDR is among the top regulators of genes induced by SARS-CoV2. Importantly, genes normally down-regulated by VitD were de-repressed in CD4+ BALF T cells of COVID-19, indicating that the VitD-driven shutdown program is impaired in this setting. The active metabolite of VitD, alfacalcidol, and cortico-steroids were among the top predicted pharmaceuticals that could normalize SARS-CoV2 induced genes. These data indicate that adjunct therapy with VitD in the context of other immunomodulatory drugs may be a beneficial strategy to dampen hyperinflammation in severe COVID-19.
- Published
- 2020
30. Functional and structural analysis of cytokine-selective IL6ST defects that cause recessive hyper-IgE syndrome
- Author
-
Freia Krause, Jessica Gold, Elizabeth A. Worthey, Diane B. Zastrow, Euan A. Ashley, Colleen E. McCormack, Michael D. W. Griffin, Dirk Schmidt-Arras, Michael W. Parker, Arian Laurence, Paul G. Fisher, Christine M. Eng, Stephen B. Montgomery, Veerabahu Shanmugasundaram, Tracy L Putoczki, Yin-Huai Chen, Shruti Marwaha, Lisa Gartner, Riley D. Metcalfe, David P. Bick, Laure Fresard, Yong Huang, Jonathan A. Bernstein, Holm H. Uhlig, Craig J. Morton, Chunli Zhao, and Matthew T. Wheeler
- Subjects
0301 basic medicine ,Male ,Immunology ,Mutation, Missense ,Genes, Recessive ,Molecular Dynamics Simulation ,Compound heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Exome Sequencing ,Cytokine Receptor gp130 ,Immunology and Allergy ,Humans ,RNA-Seq ,Child ,Exome ,Exome sequencing ,Genetics ,biology ,Oncostatin M ,Oncostatin M receptor ,Glycoprotein 130 ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cytokines ,Cytokine receptor ,Leukemia inhibitory factor ,Job Syndrome ,Signal Transduction - Abstract
Background Biallelic variants in IL6ST, encoding GP130, cause a recessive form of hyper-IgE syndrome (HIES) characterized by high IgE level, eosinophilia, defective acute phase response, susceptibility to bacterial infections, and skeletal abnormalities due to cytokine-selective loss of function in GP130, with defective IL-6 and IL-11 and variable oncostatin M (OSM) and IL-27 levels but sparing leukemia inhibitory factor (LIF) signaling. Objective Our aim was to understand the functional and structural impact of recessive HIES-associated IL6ST variants. Methods We investigated a patient with HIES by using exome, genome, and RNA sequencing. Functional assays assessed IL-6, IL-11, IL-27, OSM, LIF, CT-1, CLC, and CNTF signaling. Molecular dynamics simulations and structural modeling of GP130 cytokine receptor complexes were performed. Results We identified a patient with compound heterozygous novel missense variants in IL6ST (p.Ala517Pro and the exon-skipping null variant p.Gly484_Pro518delinsArg). The p.Ala517Pro variant resulted in a more profound IL-6– and IL-11–dominated signaling defect than did the previously identified recessive HIES IL6ST variants p.Asn404Tyr and p.Pro498Leu. Molecular dynamics simulations suggested that the p.Ala517Pro and p.Asn404Tyr variants result in increased flexibility of the extracellular membrane–proximal domains of GP130. We propose a structural model that explains the cytokine selectivity of pathogenic IL6ST variants that result in recessive HIES. The variants destabilized the conformation of the hexameric cytokine receptor complexes, whereas the trimeric LIF-GP130-LIFR complex remained stable through an additional membrane-proximal interaction. Deletion of this membrane-proximal interaction site in GP130 consequently caused additional defective LIF signaling and Stuve-Wiedemann syndrome. Conclusion Our data provide a structural basis to understand clinical phenotypes in patients with IL6ST variants.
- Published
- 2020
31. A variant in IL6ST with a selective IL-11 signaling defect in human and mouse
- Author
-
Dirk Schmidt-Arras, S Manrique, Jürgen Scheller, Glüer C-C., Jonathan Jung, Arian Laurence, Wilkie Aom., Dominik Aschenbrenner, Steven A. Wall, Miryam Müller, Chen Y-H., U Borgmeyer, T Damm, Twigg Srf., Neele Schumacher, F Krause, E Y Jones, Stefan Rose-John, Tobias Schwerd, and Holm H. Uhlig
- Subjects
0301 basic medicine ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Pathogenesis ,Ciliary neurotrophic factor ,lcsh:Physiology ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Bone ,Receptor ,lcsh:QH301-705.5 ,Phenocopy ,lcsh:QP1-981 ,biology ,Transfection ,Glycoprotein 130 ,Penetrance ,Cell biology ,030104 developmental biology ,Cytokine ,lcsh:Biology (General) ,biology.protein ,Cytokine receptor ,030217 neurology & neurosurgery - Abstract
The GP130 cytokine receptor subunit encoded by IL6ST is the shared receptor for ten cytokines of the IL-6 family. We describe a homozygous non-synonymous variant in IL6ST (p.R281Q) in a patient with craniosynostosis and retained deciduous teeth. We characterize the impact of the variant on cytokine signaling in vitro using transfected cell lines as well as primary patient-derived cells and support these findings using a mouse model with the corresponding genome-edited variant Il6st p.R279Q. We show that human GP130 p.R281Q is associated with selective loss of IL-11 signaling without affecting IL-6, IL-27, OSM, LIF, CT1, CLC, and CNTF signaling. In mice Il6st p.R279Q lowers litter size and causes facial synostosis and teeth abnormalities. The effect on IL-11 signaling caused by the GP130 variant shows incomplete penetrance but phenocopies aspects of IL11RA deficiency in humans and mice. Our data show that a genetic variant in a pleiotropic cytokine receptor can have remarkably selective defects.
- Published
- 2020
- Full Text
- View/download PDF
32. SARS-CoV2 drives JAK1/2-dependent local and systemic complement hyper-activation
- Author
-
Arian Laurence, Shahram Kordasti, Matthew R. Olson, Didier Portilla, Daniel Chauss, Luopin Wang, Claudia Kemper, Majid Kazemian, Michail S. Lionakis, Jack A. Bibby, Bingyu Yan, Erin E. West, Behdad Afzali, and Tilo Freiwald
- Subjects
Myeloid ,biology ,medicine.diagnostic_test ,Angiotensin-converting enzyme ,Regulome ,Virus ,Article ,Complement system ,medicine.anatomical_structure ,Bronchoalveolar lavage ,Interferon ,medicine ,biology.protein ,Cancer research ,Receptor ,medicine.drug - Abstract
Patients with coronavirus disease 2019 (COVID-19) present with a range of devastating acute clinical manifestations affecting the lungs, liver, kidneys and gut. The best-characterized entry receptor for the disease-causing virus SARS-CoV2, angiotensin converting enzyme (ACE) 2, is highly expressed in these tissues. However, the pathways that underlie the disease are still poorly understood. Here we show that the complement system is unexpectedly one of the intracellular pathways most highly induced by SARS-CoV2 infection in lung epithelial and liver cells. Within cells of the bronchoalveolar lavage of patients, distinct signatures of complement activation in myeloid, lymphoid and epithelial cells tracked with disease severity. Modelling the regulome of host genes induced by COVID-19 and the drugs that could normalize these genes both implicated the JAK1/2-STAT1 signaling system downstream of type I interferon receptors, and NF-kB. Ruxolitinib, a JAK1/2 inhibitor and the top predicted pharmaceutical candidate, normalized interferon signature genes, IL-6 (the best characterized severity marker in COVID-19) and all complement genes induced by SARS-CoV2, but did not affect NF-kB-regulated genes. We predict that combination therapy with JAK inhibitors and other agents with the potential to normalize NF-kB-signaling, such as anti-viral agents, may serve as an effective clinical strategy.
- Published
- 2020
33. Dominant-negative mutations in human IL6ST underlie hyper-IgE syndrome
- Author
-
Vivien, Béziat, Simon J, Tavernier, Yin-Huai, Chen, Cindy S, Ma, Marie, Materna, Arian, Laurence, Jens, Staal, Dominik, Aschenbrenner, Lisa, Roels, Lisa, Worley, Kathleen, Claes, Lisa, Gartner, Lisa A, Kohn, Marieke, De Bruyne, Klaus, Schmitz-Abe, Louis-Marie, Charbonnier, Sevgi, Keles, Justine, Nammour, Natasha, Vladikine, Majistor Raj Luxman, Maglorius Renkilaraj, Yoann, Seeleuthner, Mélanie, Migaud, Jérémie, Rosain, Mohamed, Jeljeli, Bertrand, Boisson, Eva, Van Braeckel, Jill A, Rosenfeld, Hongzheng, Dai, Lindsay C, Burrage, David R, Murdock, Bart N, Lambrecht, Véronique, Avettand-Fenoel, Tiphanie P, Vogel, Charles R, Esther, Sule, Haskologlu, Figen, Dogu, Peter, Ciznar, David, Boutboul, Marie, Ouachée-Chardin, Jean, Amourette, Marie-Noëlle, Lebras, Clément, Gauvain, Colas, Tcherakian, Aydan, Ikinciogullari, Rudi, Beyaert, Laurent, Abel, Joshua D, Milner, Bodo, Grimbacher, Louis-Jean, Couderc, Manish J, Butte, Alexandra F, Freeman, Émilie, Catherinot, Claire, Fieschi, Talal A, Chatila, Stuart G, Tangye, Holm H, Uhlig, Filomeen, Haerynck, Jean-Laurent, Casanova, Anne, Puel, St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University [New York], Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Laboratory of Human Genetics of Infectious Diseases (Necker Branch - INSERM U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Ghent University Hospital, VIB-UGent Center for Inflammation Research [Gand, Belgique] (IRC), VIB [Belgium], Center for Medical Genetics [Ghent], John Radcliffe Hospital [Oxford University Hospital], University of Oxford [Oxford], Garvan Institute of Medical Research [Darlinghurst, Australia], St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales [Sydney] (UNSW), University of California, Boston Children's Hospital, Harvard Medical School [Boston] (HMS), Necmettin Erbakan University [Konya, Turquie], Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Baylor College of Medicine (BCM), Baylor University, Laboratoire de Virologie [CHU Necker], CHU Necker - Enfants Malades [AP-HP], University of North Carolina [Chapel Hill] (UNC), University of North Carolina System (UNC), Ankara University School of Medicine [Turkey], Comenius University in Bratislava, Service d'Immunopathologie [Hôpital Saint-Louis, Paris], Hopital Saint-Louis [AP-HP] (AP-HP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université de Paris (UP), Unité d'Hémato-Immunologie pédiatrique [Hôpital Robert Debré, Paris], Service d'Immuno-hématologie pédiatrique [Hôpital Robert Debré, Paris], Hôpital Robert Debré-Hôpital Robert Debré, Groupe Hospitalier Artois-Ternois Centre Hospitalier d’Arras, Service de pneumologie [Hôpital Foch], Hôpital Foch [Suresnes], National Institute of Allergy and Infectious Diseases [Bethesda] (NIAID-NIH), National Institutes of Health [Bethesda] (NIH), Columbia University Irving Medical Center (CUIMC), Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany, Centre for Integrative Biological Signaling Studies [Freiburg, Germany], University of Freiburg [Freiburg], Hanover Medical School, Laboratoire de recherche sur les mécanismes moléculaires et pharmacologiques de l’obstruction bronchique (LOBIP), Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université de Versailles Saint-Quentin-en-Yvelines - UFR Sciences de la santé Simone Veil (UVSQ Santé), Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, Institut de Recherche Saint-Louis - Hématologie Immunologie Oncologie (Département de recherche de l’UFR de médecine, ex- Institut Universitaire Hématologie-IUH) (IRSL), Université de Paris (UP), St. Vincent’s Clinical School [Sydney, Australia], UNSW Faculty of Medicine [Sydney], and University of New South Wales [Sydney] (UNSW)-University of New South Wales [Sydney] (UNSW)
- Subjects
Male ,Adolescent ,Models, Biological ,Young Adult ,Th2 Cells ,Loss of Function Mutation ,Cytokine Receptor gp130 ,Humans ,Immunodeficiency ,Child ,Alleles ,Cells, Cultured ,Genes, Dominant ,Human disease genetics ,Cell Membrane ,Correction ,Fibroblasts ,Middle Aged ,Pedigree ,Up-Regulation ,Kinetics ,C-Reactive Protein ,Genetics, Population ,HEK293 Cells ,Phenotype ,Mutation ,Cytokines ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Female ,Job Syndrome - Abstract
International audience; Autosomal dominant hyper-IgE syndrome (AD-HIES) is typically caused by dominant-negative (DN) STAT3 mutations. Patients suffer from cold staphylococcal lesions and mucocutaneous candidiasis, severe allergy, and skeletal abnormalities. We report 12 patients from 8 unrelated kindreds with AD-HIES due to DN IL6ST mutations. We identified seven different truncating mutations, one of which was recurrent. The mutant alleles encode GP130 receptors bearing the transmembrane domain but lacking both the recycling motif and all four STAT3-recruiting tyrosine residues. Upon overexpression, the mutant proteins accumulate at the cell surface and are loss of function and DN for cellular responses to IL-6, IL-11, LIF, and OSM. Moreover, the patients’ heterozygous leukocytes and fibroblasts respond poorly to IL-6 and IL-11. Consistently, patients with STAT3 and IL6ST mutations display infectious and allergic manifestations of IL-6R deficiency, and some of the skeletal abnormalities of IL-11R deficiency. DN STAT3 and IL6ST mutations thus appear to underlie clinical phenocopies through impairment of the IL-6 and IL-11 response pathways.
- Published
- 2020
- Full Text
- View/download PDF
34. ATP6V0d2 Suppresses Alveoli Macrophage Alternative Polarization and Allergic Asthma via Degradation of PU.1
- Author
-
Xiang-Ping Yang, Arian Laurence, Na Liu, Guohua Zhen, Xiang Cheng, Zheng Liu, Zhaohui Tang, Yuchen Feng, Huicheng Liu, Wenliang Wu, Min Wu, Huabin Li, Zhengping Wei, Junyan Han, Yuxia Liang, and Pingfei Li
- Subjects
Pulmonary and Respiratory Medicine ,Protein subunit ,Immunology ,V-type ATPase ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immunology and Allergy ,V-ATPase ,CCL17 ,030223 otorhinolaryngology ,Asthma ,biology ,business.industry ,respiratory system ,medicine.disease ,respiratory tract diseases ,Ovalbumin ,030228 respiratory system ,Pu.1 protein ,Alveolar macrophage ,biology.protein ,Sputum ,Original Article ,medicine.symptom ,alveolar macrophage ,business - Abstract
Purpose Macrophages are important regulators of environmental allergen-induced airway inflammation and asthma. ATP6V0d2 is a subunit of vacuolar ATPase highly expressed in macrophages. However, the functions of ATP6V0d2 in the regulation of pathogenesis of allergic asthma remain unclear. The aim of this study is to determine the function and related molecular mechanisms of macrophage protein ATP6V0d2 in allergic asthma. Methods We compared the disease severity between female C57BL/6 wild-type and ATP6V0d2-/- mice in an ovalbumin (OVA)-induced asthma model. We also investigated the association of expression of ATP6V0d2, PU.1 and CCL17 with disease severity among asthmatic patients. Results The expression of ATP6V0d2 in sputum cells of asthmatic patients and in the lungs of OVA-challenged mice was enhanced compared to healthy subjects and their counterparts, respectively. However, ATP6V0d2-deficient mice exaggerated inflammatory cell infiltration as well as enhanced alternative activated macrophage (AAM) polarization and mucus production in an OVA-induced asthma model. Furthermore, we found that Atp6v0d2 promoted lysosomal degradation of Pu.1, which induced AAM polarization and Ccl17 production. Among asthma patients, ATP6V0d2 expression was inversely associated with disease severity, whereas PU.1 and CCL17 expression was positively associated with disease severity. Conclusions Our results identify macrophage Atp6v0d2, as an induced feedback inhibitor of asthma disease severity by promoting Pu.1 lysosomal degradation, which may in turn leads to reduced AAM polarization and Ccl17 production.
- Published
- 2020
35. Complement activates an autocrine Vitamin D system that recruits a defined transcription factor network to shut down pro-inflammatory programs of Th1 cells
- Author
-
Daniel C Chauss, Tilo Freiwald, Reuben McGregor, Bingyu Yan, Luopin Wang, Estafania Nova-Lamperti, Dhaneshwar Kumar, Zonghao Zhang, Heather Teague, Erin West, Kevin M Vannella, Marcos J Ramos-Benitez, Jack Bibby, Audrey Kelly, Amna Malik, Alexandra F Freeman, Daniella M Schwartz, Didier Portilla, Daniel S Chertow, Susan John, Paul Lavender, Claudia Kemper, Giovanna Lombardi, Nehal N Mehta, Nichola Cooper, Michail S Lionakis, Arian Laurence, Majid Kazemian, and Behdad Afzali
- Subjects
Immunology ,Immunology and Allergy - Abstract
Background Pro-inflammatory CD4+ T helper (Th)1 cells clear pathogens effectively but cause excessive tissue injury if not shut down appropriately. The complement (C’) system both induces Th1 differentiation and their shutdown, but the mechanisms regulating orderly shutdown remain unknown. Hypothesis C’ receptor engagement recruits transcriptional regulators essential to Th1 shutdown. Methods Multi-modal profiling of activated, or patient-derived Th cells, psoriatic skin, and SARS-CoV2-infected tissues was carried out by epigenome profiling, RNAseq, network modeling, phospho-arrays, confocal, and regulator knockdown. Results C’ receptor signaling induced the vitamin D (VitD) receptor (VDR) and CYP27B1, the enzyme that activates VitD, allowing T cells to both fully activate and respond to VitD. Active VitD shut down IFN-γ production by Th1 cells and induced IL-10. This was mediated by activation of IL-6 production by T cells and signaling through STAT3. Mechanistically, VitD reprogrammed the Th1 transcriptomes by forming super-enhancers and recruiting a transcription factor (TF) network consisting of VDR, c-JUN, STAT3, and BACH2. We mapped genome-wide targets of these TFs by CUT&RUN/Tag. As proof of principal, psoriatic skin treated with VitD induced BACH2 in Th cells, and genetic deficiency of either BACH2 or STAT3 inhibited IL-10 produced in response to VitD. Bronchoalveolar lavage fluid of COVID-19 patients, a C’-rich environment, showed excessive Th1 skewing and perturbation of the VitD-regulated program of genes. Conclusion We identified a C’-recruited autocrine VitD system as key to Th1 shutdown and indicate the potential for adjunct therapy with VitD in hyper-inflammatory syndromes, e.g. COVID-19. This work was supported by the Wellcome Trust (grant 097261/Z/11/Z to B.A.), the Crohn’s and Colitis Foundation of America (grant CCFA no. 3765 — CCFA genetics initiative to A.L.), British Heart Foundation (grant RG/13/12/30395 to G.L.), the National Institute of General Medical Sciences (R35GM138283 to M.K.), the Showalter Trust (research award to M.K.), German Research Foundation (DFG scholarship to T.F.; FR 3851/2-1), the NIDDK (DK12262401A1 to D.P.) and the National Agency of Research and Development of Chile (grant PAI79170073 to E.N.L.). Research was also supported by the National Institute for Health Research (NIHR) Biomedical Research Centre based at Guy’s and St Thomas’ NHS Foundation Trust and King’s College London and/or the NIHR Clinical Research Facility. The views expressed are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health. This research was supported (in part) by the Intramural Research Programs of the NIDDK (project no. ZIA/DK075149 to B.A), the National Heart, Lung and Blood Institute (project nos. ZIA/Hl006223 to C.K. and ZIA/HL006193 to N.M.), the NIAID (project no. ZIA/AI001175 to M.S.L.) of the NIH. D.C. is supported by an NIH Office of Dietary Supplements research scholar award.
- Published
- 2022
- Full Text
- View/download PDF
36. TNF overproduction impairs epithelial staphylococcal response in hyper IgE syndrome
- Author
-
Cedar J. Fowler, Ian N. Moore, Alexandra F. Freeman, Dirk A. Darnell, Mary Garofalo, Mark D. Kieh, Steven M. Holland, Pamela Welch, Arian Laurence, Sundar Ganesan, Portia Gough, John I. Gallin, Kelli W. Williams, Erik D. Anderson, Noah J. Earland, Arhum Saleem, Sandip K. Datta, Ian A. Myles, Inka Sastalla, Kol A. Zarember, and Douglas B. Kuhns
- Subjects
Adult ,Keratinocytes ,Male ,0301 basic medicine ,Staphylococcus aureus ,Epithelial-Mesenchymal Transition ,medicine.medical_treatment ,Mice, Transgenic ,medicine.disease_cause ,Immunoglobulin E ,Mice ,03 medical and health sciences ,Immune system ,medicine ,Animals ,Humans ,STAT3 ,biology ,Tumor Necrosis Factor-alpha ,business.industry ,Interleukin-17 ,Furunculosis ,Epithelial Cells ,General Medicine ,Immunotherapy ,Epithelium ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,biology.protein ,Female ,Tumor necrosis factor alpha ,Keratinocyte ,business ,Job Syndrome ,Research Article - Abstract
Autosomal dominant hyper IgE syndrome (AD-HIES), or Job's syndrome, is a primary immune deficiency caused by dominant-negative mutations in STAT3. Recurrent Staphylococcus aureus skin abscesses are a defining feature of this syndrome. A widely held hypothesis that defects in peripheral Th17 differentiation confer this susceptibility has never been directly evaluated. To assess the cutaneous immune response in AD-HIES, we induced suction blisters in healthy volunteers (HVs) and patients with AD-HIES and then challenged the wound with lethally irradiated bacteria. We show that cutaneous production of IL-17A and IL-17F was normal in patients with AD-HIES. Overproduction of TNF-α differentiated the responses in AD-HIES from HVs. This was associated with reduced IL-10 family signaling in blister-infiltrating cells and defective epithelial cell function. Mouse models of AD-HIES recapitulated these aberrant epithelial responses to S. aureus and involved defective epithelial-to-mesenchymal transition (EMT) rather than a failure of bacterial killing. Defective responses in mouse models of AD-HIES and primary keratinocyte cultures from patients with AD-HIES could be reversed by TNF-α blockade and by drugs with reported modulatory effects on EMT. Our results identify these as potential therapeutic approaches in patients with AD-HIES suffering S. aureus infections.
- Published
- 2018
- Full Text
- View/download PDF
37. Effect of Huaier granule on recurrence after curative resection of HCC: a multicentre, randomised clinical trial
- Author
-
Lianxin Liu, Chao-Liu Dai, Yan Chen, Xi-Hu Qin, Xiaoping Chen, L Zhang, Qiang Li, Bao-Cai Xing, Xi-Yan Wang, Yi-Jun Wang, Qi-Shun Zhang, Bao-Gang Peng, Hao Wen, Qian Chen, Yi Mu, Jingfeng Liu, Zhiren Fu, Ping Bie, Li Bo, Weiping Zhou, Xin-Yu Peng, Arian Laurence, Jian-Qiang Cai, Yi-Tao Ding, Ping Yin, Chang Shu, Ge-Liang Xu, Zhi-Wei Zhang, Zuo-Jun Zhen, Qi-Chang Zheng, Shou-Wang Cai, Yu-Bao Zhang, Le-Qun Li, Bin Jiang, Hai-Xin Qian, and Xue-Wen Zhang
- Subjects
Male ,0301 basic medicine ,Curative resection ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Complex Mixtures ,Gastroenterology ,Resection ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Clinical endpoint ,Adjuvant therapy ,Hepatectomy ,Humans ,Aged ,Trametes ,business.industry ,Liver Neoplasms ,Huaier Granule ,Middle Aged ,medicine.disease ,Survival Analysis ,Clinical trial ,Treatment Outcome ,030104 developmental biology ,Liver ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Female ,Neoplasm Recurrence, Local ,business - Abstract
ObjectiveThere is little evidence that adjuvant therapy after radical surgical resection of hepatocellular carcinoma (HCC) improves recurrence-free survival (RFS) or overall survival (OS). We conducted a multicentre, randomised, controlled, phase IV trial evaluating the benefit of an aqueous extract of Trametes robinophila Murr (Huaier granule) to address this unmet need.Design and resultsA total of 1044 patients were randomised in 2:1 ratio to receive either Huaier or no further treatment (controls) for a maximum of 96 weeks. The primary endpoint was RFS. Secondary endpoints included OS and tumour extrahepatic recurrence rate (ERR). The Huaier (n=686) and control groups (n=316) had a mean RFS of 75.5 weeks and 68.5 weeks, respectively (HR 0.67; 95% CI 0.55 to 0.81). The difference in the RFS rate between Huaier and control groups was 62.39% and 49.05% (95% CI 6.74 to 19.94; p=0.0001); this led to an OS rate in the Huaier and control groups of 95.19% and 91.46%, respectively (95% CI 0.26 to 7.21; p=0.0207). The tumour ERR between Huaier and control groups was 8.60% and 13.61% (95% CI −12.59 to −2.50; p=0.0018), respectively.ConclusionsThis is the first nationwide multicentre study, involving 39 centres and 1044 patients, to prove the effectiveness of Huaier granule as adjuvant therapy for HCC after curative liver resection. It demonstrated a significant prolongation of RFS and reduced extrahepatic recurrence in Huaier group.Trial registrationNCT01770431; Post-results.
- Published
- 2018
- Full Text
- View/download PDF
38. STAT-3–independent production of IL-17 by mouse innate-like αβ T cells controls ocular infection
- Author
-
Katrin D. Mayer-Barber, Rachel R. Caspi, Phyllis B. Silver, Cheng-Rong Yu, Charles E. Egwuagu, Reiko Horai, Arian Laurence, Hatice Karauzum, Anthony J. St. Leger, Sandip K. Datta, Rafael Villasmil, and Anna M. Hansen
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,Staphylococcus aureus ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,T cell ,Eye Infections ,Immunology ,Inflammation ,Thymus Gland ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,medicine ,Animals ,Immunology and Allergy ,Promyelocytic Leukemia Zinc Finger Protein ,Phosphorylation ,Transcription factor ,Research Articles ,Mice, Knockout ,Mucous Membrane ,Interleukins ,Interleukin-17 ,T-cell receptor ,Brief Definitive Report ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,eye diseases ,Immunity, Innate ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Th17 Cells ,Interleukin 17 ,medicine.symptom ,Signal transduction ,Immunologic Memory ,CD8 ,Signal Transduction ,030215 immunology - Abstract
St. Leger et al. identify and examine innate-like αβ T cells that circumvent canonical STAT-3 phosphorylation to produce protective IL-17. These cells can exist in the ocular mucosa and protect the ocular surface from pathogenic Staphylococcus aureus infection., Appropriate regulation of IL-17 production in the host can mean the difference between effective control of pathogens and uncontrolled inflammation that causes tissue damage. Investigation of conventional CD4+ T cells (Th17 cells) has yielded invaluable insights into IL-17 function and its regulation. More recently, we and others reported production of IL-17 from innate αβ+ T cell populations, which was shown to occur primarily via IL-23R signaling through the transcription factor STAT-3. In our current study, we identify promyelocytic leukemia zinc finger (PLZF)–expressing iNKT, CD4−/CD8+, and CD4−/CD8− (DN) αβ+T cells, which produce IL-17 in response to TCR and IL-1 receptor ligation independently of STAT-3 signaling. Notably, this noncanonical pathway of IL-17 production may be important in mucosal defense and is by itself sufficient to control pathogenic Staphylococcus aureus infection at the ocular surface., Graphical Abstract
- Published
- 2018
- Full Text
- View/download PDF
39. Multi-organ graft-versus-host disease after nivolumab for relapsed Hodgkin lymphoma: the role of plasma exchange
- Author
-
Harpreet Hyare, Maeve A O'Reilly, Karl S. Peggs, Rodothea Amerikanou, Harriet Roddy, Arian Laurence, Kirsty Thomson, Sian Hughes, Manar S. Shafat, Lorna Neill, and Claire Roddie
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Adolescent ,Plasma Exchange ,business.industry ,medicine.medical_treatment ,Hematopoietic Stem Cell Transplantation ,Graft vs Host Disease ,Hematology ,Hematopoietic stem cell transplantation ,Multi organ ,medicine.disease ,Hodgkin Disease ,Antineoplastic Agents, Immunological ,Nivolumab ,Neoplasm Recurrence ,Graft-versus-host disease ,Internal medicine ,medicine ,Humans ,Hodgkin lymphoma ,Neoplasm Recurrence, Local ,business - Published
- 2021
- Full Text
- View/download PDF
40. Targeting JAK/STAT signalling in inflammatory skin diseases with small molecule inhibitors
- Author
-
Arian Laurence, Katharina Welsch, Kamran Ghoreschi, and Julia Holstein
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Autoimmunity ,Inflammation ,medicine.disease_cause ,Skin Diseases ,stat ,03 medical and health sciences ,Psoriasis ,medicine ,Humans ,Immunology and Allergy ,Receptors, Cytokine ,Receptor ,Protein Kinase Inhibitors ,Janus Kinases ,biology ,business.industry ,JAK-STAT signaling pathway ,medicine.disease ,STAT Transcription Factors ,030104 developmental biology ,Cytokine ,biology.protein ,Cytokines ,Antibody ,medicine.symptom ,business ,Immunosuppressive Agents ,Signal Transduction - Abstract
For most inflammatory skin diseases topical glucocorticosteroids and traditional oral immunosuppressive drugs remain the principle treatment choices, but this has started to change. A deeper understanding in individual disease pathogenesis, basic immune mechanisms and molecular signalling pathways, together with advances in pharmaceutical drug development, allow us to interfere more precisely with disease-related factors. Some examples of inflammation-controlling interventions include antibodies neutralizing disease-associated cytokines, and small molecules targeting intracellular pathways relevant to cytokine production or cytokine signalling. So far, this is best established for psoriasis, an inflammatory skin disease dominated by Th17 cytokines. In this review, we focus on chronic inflammatory skin diseases where cytokines using type I/II cytokine receptors play a dominant role in disease pathogenesis and where novel treatments with inhibitors of the JAK/STAT pathway are already under clinical investigation. To better understand the rationale of using JAK/STAT inhibitors in the discussed skin diseases, we give an overview of important genetic and immunological associations with the JAK/STAT pathway and summarize the stage of clinical development of small molecular inhibitors. JAK/STAT inhibitors will presumably find wide application in dermatology, since they can be applied not only systematically but also topically for the treatment of inflammatory skin diseases.
- Published
- 2017
- Full Text
- View/download PDF
41. List of Contributors
- Author
-
Jakub Abramson, S. Sohail Ahmed, Marco A. Alba, Youssif M. Ali, Julian L. Ambrus, Agnes Andersson Svärd, Martin Aringer, Shervin Assassi, Thanda Aung, Ilya Ayzenberg, Robert N. Barker, Alan G. Baxter, Corrado Betterle, Stanca A. Birlea, Niklas K. Björkström, Paul A. Blair, Stephan Blüml, Xavier Bosch, Robert A. Brodsky, Yenan T. Bryceson, Patrick R. Burkett, James B. Bussel, Roberto Caricchio, Livia Casciola-Rosen, Patrizio Caturegli, Benjamin Chaigne-Delalande, Paulina Chalan, Lucienne Chatenoud, Philip L. Cohen, Megan A. Cooper, Ken Coppieters, Ronald G. Crystal, Donna A. Culton, Valentina Damato, Anne Davidson, Lorenzo Delfino, Peter J. Delves, Giulia Di Dalmazi, Betty Diamond, Luis A. Diaz, Ronald J. Falk, Marvin J. Fritzler, Stefania Gallucci, Sapna Gangaputra, Brian Gelbman, M. Eric Gershwin, Igal Gery, Daniel R. Getts, Ralf Gold, Yael Goldfarb, Jing Gong, Siamon Gordon, Jörg J. Goronzy, Judith M. Greer, Vanesa A. Guazzone, Luiza Guilherme, David A. Hafler, Bevra H. Hahn, Abdel Rahim A. Hamad, Hideaki Hamano, Leonard C. Harrison, Dirk Homann, Eystein S. Husebye, J. Charles Jennette, Richard J. Jones, Margaret A. Jordan, Jorge Kalil, Shigeyuki Kawa, Ziya Kaya, Christian W. Keller, Nicholas J.C. King, Maleewan Kitcharoensakkul, Kendo Kiyosawa, Christoph Königs, Mitchell Kronenberg, Vijay K. Kuchroo, Arian Laurence, Eun-Ju Lee, Helmar C. Lehmann, Åke Lernmark, Ida Lindbladh, Zhi Liu, Hans-Gustaf Ljunggren, Claudio Lunardi, Knut E.A. Lundin, Jan D. Lünemann, Michael P.T. Lunn, Livia Lustig, Charles R. Mackay, Ian R. Mackay, Clara Malattia, Luisa Martinez-Pomares, Alberto Martini, Claudia Mauri, Pamela A. McCombe, Fritz Melchers, Giorgina Mieli-Vergani, Frederick W. Miller, Stephen D. Miller, Masayuki Mizui, Jenny Mjösberg, Christian Münz, Jagtar Singh Nijjar, David A. Norris, Kristine Oleinika, Joost J. Oppenheim, Mathias Pawlak, Cristina Peligero-Cruz, Anneli Peters, Pärt Peterson, Kalliopi Pitarokoili, Fabio Presotto, Antonio Puccetti, Hamid Rabb, Patricia Raczek, M. Jubayer Rahman, Manuel Ramos-Casals, Noel R. Rose, Antony Rosen, Mohanraj Sadasivam, Adam Schiffenbauer, Wilhelm J. Schwaeble, H. Nida Sen, Marc Serota, Kazim A. Sheikh, Yehuda Shoenfeld, Ora Shovman, Joachim Sieper, Arthur M. Silverstein, Robert B. Sim, Kenneth G C Smith, Josef S. Smolen, Ludvig M. Sollid, Alanna Spiteri, Lawrence Steinman, John H. Stone, Uta Syrbe, Ami Tamhaney, Atsushi Tanaka, Veena Taneja, Kristin V. Tarbell, Elisa Tinazzi, Benedict K. Tiong, Ban-Hock Toh, George C. Tsokos, Kenneth S.K. Tung, John Varga, Diego Vergani, Mark A. Vickers, Stuart Viegas, Angela Vincent, Matthias von Herrath, Anthony P. Weetman, Joel V. Weinstock, John M. Wentworth, Sarah Wesley, Cornelia M. Weyand, Gerhard Wingender, Michael W. Winter, Renato Zanchetta, and Moncef Zouali
- Published
- 2020
- Full Text
- View/download PDF
42. Effector Mechanisms in Autoimmunity
- Author
-
Arian Laurence and Martin Aringer
- Subjects
Autoimmune disease ,Effector ,CCL18 ,Autoantibody ,Inflammation ,Biology ,medicine.disease ,medicine.disease_cause ,Autoimmunity ,Classical complement pathway ,Immune system ,Immunology ,medicine ,medicine.symptom - Abstract
Autoimmune disease results from an inability of the immune system to differentiate self-antigens from those of a foreign and harmful organism. Since the immune system is forced to deal with an unforeseeable variety of pathogens, it is impossible to design an effective immune system that is incapable of reacting against self-antigens. Instead, a complex system of checks and balances has evolved to keep the immune system from reacting harmfully against “self.” Accordingly, while autoimmune T cells and B cells may well be part of the healthy immune system, as often are autoantibodies, a failure to repress these cells results in the activation of the immune system’s effector mechanisms. This will almost always lead to inflammation and damage, i.e., autoimmune disease. In the absence of a reliable way of specifically removing autoimmune cells, most successful clinical strategies target the effector mechanisms in the treatment of autoimmunity. It is therefore important to understand these effector mechanisms and to both recognize their role in autoimmune diseases and minimize their harmful effects.
- Published
- 2020
- Full Text
- View/download PDF
43. Death-associated protein kinase 1 (DAPK1) controls CD8
- Author
-
Zhengping, Wei, Qiuyang, Du, Pingfei, Li, Huicheng, Liu, Minghui, Xia, Yufei, Chen, Guoyu, Bi, Zhao-Hui, Tang, Xiang, Cheng, Youming, Lu, Ran, He, Arian, Laurence, Jing, Wang, Liu, Huang, Huabin, Li, and Xiang-Ping, Yang
- Subjects
Mice, Knockout ,Death-Associated Protein Kinases ,Immunity, Cellular ,Mice ,Cell Movement ,Cell Line, Tumor ,Animals ,Neoplasms, Experimental ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation - Abstract
Appropriate migration of cytotoxic T effector cells into the tumors is crucial for their antitumor function. Despite the controversial role of PI3K-Akt in CD8
- Published
- 2019
44. RNF144A shapes the hierarchy of cytokine signaling to provide protective immunity against influenza
- Author
-
N. Merle, Dragana Jankovic, N. Cheru, Suman Mitra, Arian Laurence, Zu-Xi Yu, Hiroyuki Nagashima, Claudia Kemper, Alejandro V. Villarino, Majid Kazemian, Susan D. John, M. Bijlmakers, Estefania Nova-Lamperti, John J. O'Shea, Bingyu Yan, Erin E. West, Behdad Afzali, Gregory Weitsman, Daniel Chauss, S. Kim, and Tilo Freiwald
- Subjects
0303 health sciences ,biology ,Effector ,medicine.medical_treatment ,3. Good health ,Cell biology ,Ubiquitin ligase ,03 medical and health sciences ,0302 clinical medicine ,Cytokine ,Immune system ,Granzyme ,biology.protein ,medicine ,Tumor necrosis factor alpha ,Signal transduction ,030217 neurology & neurosurgery ,STAT5 ,030304 developmental biology - Abstract
Cytokine-induced signaling pathways are tightly regulated and self-limiting, as their dysregulation causes immune disorders and cancer. The precise mechanisms that fine-tune these responses are incompletely understood. We show that the E3 ubiquitin ligaseRNF144Ais an IL-2/STAT5-induced gene in T cells and critically orchestrates the hierarchy of IL-2R signaling to promote STAT5 activation and limit RAF-ERK-MAPK output from the IL-2R. Mechanistically, RNF144A increased the interaction between IL-2Rβ and STAT5 and polyubiquitinated RAF1, enhancing its proteasomal degradation and preventing the formation of the potent RAF1/BRAF kinase complex. CD8+T cells fromRnf144a–/–mice had impaired IL-2-induction of effector genes, includingTnfand granzymes, and these mice demonstrated increased susceptibility to influenza. Reduced RNF144A expression was associated with more severe influenza in humans and its expression in patients was a biomarker distinguishing moderate from severe disease. These studies reveal a vital physiological role for RNF144A in maintaining the fidelity of IL-2R signaling in CTLs to prevent severe inflammation in response to infection.One Sentence SummaryRNF144A promotes anti-viral immunity by regulating the hierarchy of cytokine signal output
- Published
- 2019
- Full Text
- View/download PDF
45. DAPK1 (death associated protein kinase 1) mediates mTORC1 activation and antiviral activities in CD8(+) T cells
- Author
-
Guoyu Bi, Huicheng Liu, Ran He, Minghui Xia, Na Liu, Arian Laurence, Zhaohui Tang, Yu Xia, Zhuoya Li, Lilin Ye, Pingfei Li, Xiang Cheng, Youming Lu, Qiuyang Du, Lei Pei, Guihua Wang, Zhengping Wei, Xiang-Ping Yang, Huabin Li, and Jing Wang
- Subjects
0301 basic medicine ,Immunology ,mTORC1 ,CD8-Positive T-Lymphocytes ,Mechanistic Target of Rapamycin Complex 1 ,Lymphocyte Activation ,Antiviral Agents ,Article ,03 medical and health sciences ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Arenaviridae Infections ,Lymphocytic choriomeningitis virus ,Kinase activity ,Phosphorylation ,PI3K/AKT/mTOR pathway ,Death domain ,Mice, Knockout ,Chemistry ,TOR Serine-Threonine Kinases ,Cell Differentiation ,Cell biology ,Mice, Inbred C57BL ,Death-Associated Protein Kinases ,030104 developmental biology ,Infectious Diseases ,Protein kinase domain ,Death-Associated Protein Kinase 1 ,biological phenomena, cell phenomena, and immunity ,030215 immunology ,Signal Transduction - Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) regulates CD8(+) T-cell differentiation and function. Despite the links between PI3K-AKT and mTORC1 activation in CD8(+) T cells, the molecular mechanism underlying mTORC1 activation remains unclear. Here, we show that both the kinase activity and the death domain of DAPK1 are required for maximal mTOR activation and CD8(+) T-cell function. We found that TCR-induced activation of calcineurin activates DAPK1, which subsequently interacts with TSC2 via its death domain and phosphorylates TSC2 to mediate mTORC1 activation. Furthermore, both the kinase domain and death domain of DAPK1 are required for CD8(+) T-cell antiviral responses in an LCMV infection model. Together, our data reveal a novel mechanism of mTORC1 activation that mediates optimal CD8(+) T-cell function and antiviral activity.
- Published
- 2019
46. The role of PTEN in innate and adaptive immunity
- Author
-
Henry Taylor, Holm H. Uhlig, and Arian Laurence
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Adaptive Immunity ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Autoimmunity ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Tensin ,PTEN ,Receptor ,PI3K/AKT/mTOR pathway ,PTEN Phosphohydrolase ,Acquired immune system ,Immunity, Innate ,030104 developmental biology ,Cytokine ,Cancer research ,biology.protein ,Hamartoma Syndrome, Multiple ,Signal Transduction ,Perspectives ,030215 immunology - Abstract
The lipid and protein phosphatase and tensin homolog (PTEN) controls the differentiation and activation of multiple immune cells. PTEN acts downstream from T- and B-cell receptors, costimulatory molecules, cytokine receptors, integrins, and also growth factor receptors. Loss of PTEN activity in human and mice is associated with cellular and humoral immune dysfunction, lymphoid hyperplasia, and autoimmunity. Although most patients with PTEN hamartoma tumor syndrome (PHTS) have no immunological symptoms, a subclinical immune dysfunction is present in many, and clinical immunodeficiency in few. Comparison of the immune phenotype caused by PTEN haploinsufficiency in PHTS, phosphoinositide 3-kinase (PI3K) gain-of-function in activated PI3K syndrome, and mice with conditional biallelic Pten deletion suggests a threshold model in which coordinated activity of several phosphatases control the PI3K signaling in a cell-type-specific manner. Emerging evidence highlights the role of PTEN in polygenic autoimmune disorders, infection, and the immunological response to cancer. Targeting the PI3K axis is an emerging therapeutic avenue.
- Published
- 2019
47. Protein Kinase Antagonists in Therapy of Immunological and Inflammatory Diseases
- Author
-
Arian Laurence, Massimo Gadina, and John J. O'Shea
- Subjects
Severe combined immunodeficiency ,medicine.anatomical_structure ,Immune system ,Kinase ,Mutant protein ,Cell ,medicine ,Cancer research ,Protein phosphorylation ,Biology ,Signal transduction ,medicine.disease ,Protein kinase A - Abstract
Reversible protein phosphorylation is a critical regulator of homeostasis in all eukaryotic cells. It is a major means for transmitting information from outside the cell and between the subcellular components within the cell. The importance of protein phosphorylation is illustrated both in cancer, in which mutant protein kinases function as oncogenes, and in disorders of the immune system, where inherited deficiencies of protein kinase signaling are responsible for the majority of cases of severe combined immunodeficiency. On the basis of these findings, targeting protein kinases has been proposed to be a useful strategy in the development of novel immunosuppressant drugs and is one of the most active areas of pharmaceutical drug development, with much of the impetus coming from oncology. This chapter will describe many of the key signaling pathways in immune cells and highlight attempts to design specific inhibitors of key kinases within each pathway to design drugs that limit discrete components of the immune system responsible for particular autoimmune diseases while preserving defense against infections.
- Published
- 2019
- Full Text
- View/download PDF
48. The macrophage-specific V-ATPase subunit ATP6V0D2 restricts inflammasome activation and bacterial infection by facilitating autophagosome-lysosome fusion
- Author
-
Xiaofei Deng, Kan Jiang, Jinxia Zhang, Jing Wang, Binjiao Yin, Feili Gong, John J. O'Shea, Yu Xia, Jae Jin Chae, Hongping Ba, Arian Laurence, Na Liu, Zhuoya Li, Yongwon Choi, Xiang Cheng, Yao Yao, Lin Li, Guoyu Bi, Xiuxiu Xie, Zhaohui Tang, and Xiang-Ping Yang
- Subjects
Salmonella typhimurium ,0301 basic medicine ,Vacuolar Proton-Translocating ATPases ,Inflammasomes ,Protein subunit ,Peritonitis ,Biology ,Membrane Fusion ,R-SNARE Proteins ,Mice ,03 medical and health sciences ,Autophagy ,medicine ,Animals ,Humans ,Macrophage ,V-ATPase ,Molecular Biology ,Cells, Cultured ,Adenosine Triphosphatases ,Mice, Knockout ,030102 biochemistry & molecular biology ,Qa-SNARE Proteins ,Macrophages ,Autophagosomes ,Inflammasome ,Cell Biology ,Colitis ,Autophagosome formation ,Mitochondria ,Cell biology ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Salmonella Infections ,Autophagosome lysosome fusion ,Lysosomes ,human activities ,Research Paper ,medicine.drug - Abstract
Macroautophagy/autophagy is a conserved ubiquitous pathway that performs diverse roles in health and disease. Although many key, widely expressed proteins that regulate autophagosome formation followed by lysosomal fusion have been identified, the possibilities of cell-specific elements that contribute to the autophagy fusion machinery have not been explored. Here we show that a macrophage-specific isoform of the vacuolar ATPase protein ATP6V0D2/subunit d2 is dispensable for lysosome acidification, but promotes the completion of autophagy via promotion of autophagosome-lysosome fusion through its interaction with STX17 and VAMP8. Atp6v0d2-deficient macrophages have augmented mitochondrial damage, enhanced inflammasome activation and reduced clearance of Salmonella typhimurium. The susceptibility of atp6v0d2 knockout mice to DSS-induced colitis and Salmonella typhimurium-induced death, highlights the in vivo significance of ATP6V0D2-mediated autophagosome-lysosome fusion. Together, our data identify ATP6V0D2 as a key component of macrophage-specific autophagosome-lysosome fusion machinery maintaining macrophage organelle homeostasis and, in turn, limiting both inflammation and bacterial infection. Abbreviations: ACTB/β-actin: actin, beta; ATG14: autophagy related 14; ATG16L1: autophagy related 16-like 1 (S. cerevisiae); ATP6V0D1/2: ATPase, H+ transporting, lysosomal V0 subunit D1/2; AIM2: absent in melanoma 2; BMDM: bone marrow-derived macrophage; CASP1: caspase 1; CGD: chronic granulomatous disease; CSF1/M-CSF: colony stimulating factor 1 (macrophage); CTSB: cathepsin B; DSS: dextran sodium sulfate; IL1B: interleukin 1 beta; IL6: interleukin 6; IRGM: immunity-related GTPase family M member; KO: knockout; LAMP1: lysosomal-associated membrane protein 1; LC3: microtubule-associated protein 1 light chain 3; LPS: lipo-polysaccaride; NLRP3: NLR family, pyrin domain containing 3; PYCARD/ASC: PYD and CARD domain containing; SNARE: soluble N-ethylmaleimide-sensitive factor attachment protein receptor; SNAP29: synaptosomal-associated protein 29; SQSTM1/p62: sequestosome 1; STX17: syntaxin 17; TLR: toll-like receptor; TNF: tumor necrosis factor ; TOMM20: translocase of outer mitochondrial membrane 20; ULK1: unc-51 like kinase 1; VAMP8: vesicle-associated membrane protein 8; WT: wild type; 3-MA: 3-methyladenine
- Published
- 2019
- Full Text
- View/download PDF
49. List of Contributors
- Author
-
Roshini Sarah Abraham, Cristina Albanesi, Ilias Alevizos, Juan Anguita, Brendan Antiochos, Cynthia Aranow, John P. Atkinson, Howard A. Austin, Subash Babu, Mark C. Ballow, James E. Balow, John W. Belmont, Claudia Berek, Timothy Beukelman, Tapan Bhavsar, J. Andrew Bird, Sarah E. Blutt, Mark Boguniewicz, Rafael Bonamichi-Santos, Bertrand Boisson, Elena Borzova, Prosper N. Boyaka, Joshua Boyce, Sarah K. Browne, Wesley Burks, Jacinta Bustamante, Virginia L. Calder, Matthew Campbell, Adela Rambi G. Cardones, Jean-Laurent Casanova, Mariana Castells, Lisa A. Cavacini, Edwin S.L. Chan, David D. Chaplin, W. Winn Chatham, Edward S. Chen, Javier Chinen, Lisa Christopher-Stine, Michael Ciancanelli, Andrew P. Cope, David B. Corry, Filippo Crea, Randy Q. Cron, Jennifer M. Cuellar-Rodriguez, Marinos C. Dalakas, Sara M. Dann, Betty Diamond, Terry W. Du, Stéphanie Dupuis-Boisson, Todd N. Eagar, Craig A. Elmets, Doruk Erkan, Laura Fanning, Erol Fikrig, Davide Flego, Thomas A. Fleisher, Luz Fonacier, Andrew P. Fontenot, Alexandra F. Freeman, Anthony J. Frew, Kohtaro Fujihashi, Massimo Gadina, Moshe E. Gatt, M. Eric Gershwin, Susan L. Gillespie, Jörg J. Goronzy, Sangeeta Goswami, Clive E.H. Grattan, Neil S. Greenspan, Sarthak Gupta, Claire E. Gustafson, Russell P. Hall, Robert G. Hamilton, Laurie E. Harrington, Leonard C. Harrison, Sarfaraz A. Hasni, Arthur Helbling, Joanna Hester, Steven M. Holland, Dennis Hourcade, Nicholas D. Huntington, Tracy Hwangpo, John B. Imboden, Fadi Issa, Shai Izraeli, Elaine S. Jaffe, Sirpa Jalkanen, Stacie Jones, Emmanuelle Jouanguy, Sarah Kabbani, Stefan H.E. Kaufmann, Farrah Kheradmand, Donald B. Kohn, Robert Korngold, Anna Kovalszki, Douglas B. Kuhns, Hrishikesh Kulkarni, Caroline Y. Kuo, Arash Lahouti, C. Ola Landgren, Arian Laurence, Joyce S. Lee, Catherine Lemière, Donald Y.M. Leung, Arnold I. Levinson, Ofer Levy, Dorothy E. Lewis, Phoebe Lin, Andreas Linkermann, Giovanna Liuzzo, Michael D. Lockshin, Allison K. Lord, Jay N. Lozier, Amber Luong, Raashid Luqmani, Meggan Mackay, Jonathan S. Maltzman, Peter J. Mannon, Michael P. Manns, James G. Martin, Craig L. Maynard, Samual McCash, Douglas R. McDonald, Peter C. Melby, Stephen D. Miller, Anna L. Mitchell, Amirah Mohd-Zaki, Carolyn Mold, David R. Moller, Dimitrios S. Monos, Scott N. Mueller, Catharina M. Mulders-Manders, Mark J. Mulligan, Ulrich R. Müller, Pashna N. Munshi, Kazunori Murata, Philip M. Murphy, Nicolás Navasa, Pierre Noel, Luigi D. Notarangelo, Robert L. Nussbaum, Thomas B. Nutman, Stephen L. Nutt, João B. Oliveira, Thomas L. Ortel, John J. O'Shea, Sung-Yun Pai, Lavannya Pandit, Mary E. Paul, Simon H.S. Pearce, Daniela Pedicino, Erik J. Peterson, Capucine Picard, Stefania Pittaluga, Debra Long Priel, Jennifer Puck, Anne Puel, Andreas Radbruch, Stephen T. Reece, John D. Reveille, Robert R. Rich, Chaim M. Roifman, Antony Rosen, James T. Rosenbaum, Sergio D. Rosenzweig, Barry T. Rouse, Scott D. Rowley, Shimon Sakaguchi, Marko Salmi, Andrea J. Sant, Sarah W. Satola, Valerie Saw, Marcos C. Schechter, Harry W. Schroeder, Benjamin M. Segal, Carlo Selmi, Sushma Shankar, Anu Sharma, Padmanee Sharma, William T. Shearer, Richard M. Siegel, Anna Simon, Gideon P. Smith, David S. Stephens, Robin Stephens, Alex Straumann, Leyla Y. Teos, Laura Timares, Wulf Tonnus, Raul M. Torres, Gülbü Uzel, Jeroen C.H. van der Hilst, Jos W.M. van der Meer, John Varga, Jatin M. Vyas, Meryl Waldman, Peter Weiser, Peter F. Weller, Cornelia M. Weyand, Fredrick M. Wigley, Robert J. Winchester, James B. Wing, Kathryn J. Wood, Xiaobo Wu, Hui Xu, Cassian Yee, and Shen-Ying Zhang
- Published
- 2019
- Full Text
- View/download PDF
50. Human retinoic acid-regulated CD161(+) regulatory T cells support wound repair in intestinal mucosa
- Author
-
Estefania Nova-Lamperti, Mehdi Pirooznia, Eirini Pantazi, Marco Romano, Claudia Kemper, David J. Cousins, Hong-Wei Sun, Daniel Chauss, Han-Yu Shih, Behdad Afzali, Dominic A. Boardman, Polychronis Pavlidis, Majid Kazemian, Reuben McGregor, Giovanni A M Povoleri, Arian Laurence, Pablo D. Becker, Nichola Cooper, Nick Powell, Benedetta Costantini, Giorgia Fanelli, Cristiano Scottà, Shahram Kordasti, Yun-Ching Chen, and Giovanna Lombardi
- Subjects
0301 basic medicine ,EXPRESSION ,HOMEOSTASIS ,SUBSETS ,Population ,Immunology ,Retinoic acid ,Inflammation ,chemical and pharmacologic phenomena ,Tretinoin ,INNATE ,Biology ,T-Lymphocytes, Regulatory ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,HUMAN NKR-P1A ,Intestinal mucosa ,Crohn Disease ,RAR-related orphan receptor gamma ,T-Lymphocyte Subsets ,hemic and lymphatic diseases ,medicine ,Immunology and Allergy ,FOXP3 GENE ,Humans ,Intestinal Mucosa ,education ,education.field_of_study ,Wound Healing ,Science & Technology ,INDUCTION ,SIGNATURE ,hemic and immune systems ,FOSL2 ,IN-VITRO ,EXPANSION ,030104 developmental biology ,chemistry ,1107 Immunology ,Cancer research ,medicine.symptom ,Wound healing ,Life Sciences & Biomedicine ,medicine.drug ,NK Cell Lectin-Like Receptor Subfamily B - Abstract
Repair of tissue damaged during inflammatory processes is key to the return of local homeostasis and restoration of epithelial integrity. Here we describe CD161+ regulatory T (Treg) cells as a distinct, highly suppressive population of Treg cells that mediate wound healing. These Treg cells were enriched in intestinal lamina propria, particularly in Crohn's disease. CD161+ Treg cells had an all-trans retinoic acid (ATRA)-regulated gene signature, and CD161 expression on Treg cells was induced by ATRA, which directly regulated the CD161 gene. CD161 was co-stimulatory, and ligation with the T cell antigen receptor induced cytokines that accelerated the wound healing of intestinal epithelial cells. We identified a transcription-factor network, including BACH2, RORγt, FOSL2, AP-1 and RUNX1, that controlled expression of the wound-healing program, and found a CD161+ Treg cell signature in Crohn's disease mucosa associated with reduced inflammation. These findings identify CD161+ Treg cells as a population involved in controlling the balance between inflammation and epithelial barrier healing in the gut.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.