253 results on '"Lenardo MJ"'
Search Results
2. Mutations that prevent caspase cleavage of RIPK1 cause autoinflammatory disease
- Author
-
Lalaoui, N, Boyden, SE, Oda, H, Wood, GM, Stone, DL, Chau, D, Liu, L, Stoffels, M, Kratina, T, Lawlor, KE, Zaal, KJM, Hoffmann, PM, Etemadi, N, Shield-Artin, K, Biben, C, Tsai, WL, Blake, MD, Kuehn, HS, Yang, D, Anderton, H, Silke, N, Wachsmuth, L, Zheng, L, Moura, NS, Beck, DB, Gutierrez-Cruz, G, Ombrello, AK, Pinto-Patarroyo, GP, Kueh, AJ, Herold, MJ, Hall, C, Wang, H, Chae, JJ, Dmitrieva, NI, McKenzie, M, Light, A, Barham, BK, Jones, A, Romeo, TM, Zhou, Q, Aksentijevich, I, Mullikin, JC, Gross, AJ, Shum, AK, Hawkins, ED, Masters, SL, Lenardo, MJ, Boehm, M, Rosenzweig, SD, Pasparakis, M, Voss, AK, Gadina, M, Kastner, DL, Silke, J, Lalaoui, N, Boyden, SE, Oda, H, Wood, GM, Stone, DL, Chau, D, Liu, L, Stoffels, M, Kratina, T, Lawlor, KE, Zaal, KJM, Hoffmann, PM, Etemadi, N, Shield-Artin, K, Biben, C, Tsai, WL, Blake, MD, Kuehn, HS, Yang, D, Anderton, H, Silke, N, Wachsmuth, L, Zheng, L, Moura, NS, Beck, DB, Gutierrez-Cruz, G, Ombrello, AK, Pinto-Patarroyo, GP, Kueh, AJ, Herold, MJ, Hall, C, Wang, H, Chae, JJ, Dmitrieva, NI, McKenzie, M, Light, A, Barham, BK, Jones, A, Romeo, TM, Zhou, Q, Aksentijevich, I, Mullikin, JC, Gross, AJ, Shum, AK, Hawkins, ED, Masters, SL, Lenardo, MJ, Boehm, M, Rosenzweig, SD, Pasparakis, M, Voss, AK, Gadina, M, Kastner, DL, and Silke, J
- Abstract
RIPK1 is a key regulator of innate immune signalling pathways. To ensure an optimal inflammatory response, RIPK1 is regulated post-translationally by well-characterized ubiquitylation and phosphorylation events, as well as by caspase-8-mediated cleavage1-7. The physiological relevance of this cleavage event remains unclear, although it is thought to inhibit activation of RIPK3 and necroptosis8. Here we show that the heterozygous missense mutations D324N, D324H and D324Y prevent caspase cleavage of RIPK1 in humans and result in an early-onset periodic fever syndrome and severe intermittent lymphadenopathy-a condition we term 'cleavage-resistant RIPK1-induced autoinflammatory syndrome'. To define the mechanism for this disease, we generated a cleavage-resistant Ripk1D325A mutant mouse strain. Whereas Ripk1-/- mice died postnatally from systemic inflammation, Ripk1D325A/D325A mice died during embryogenesis. Embryonic lethality was completely prevented by the combined loss of Casp8 and Ripk3, but not by loss of Ripk3 or Mlkl alone. Loss of RIPK1 kinase activity also prevented Ripk1D325A/D325A embryonic lethality, although the mice died before weaning from multi-organ inflammation in a RIPK3-dependent manner. Consistently, Ripk1D325A/D325A and Ripk1D325A/+ cells were hypersensitive to RIPK3-dependent TNF-induced apoptosis and necroptosis. Heterozygous Ripk1D325A/+ mice were viable and grossly normal, but were hyper-responsive to inflammatory stimuli in vivo. Our results demonstrate the importance of caspase-mediated RIPK1 cleavage during embryonic development and show that caspase cleavage of RIPK1 not only inhibits necroptosis but also maintains inflammatory homeostasis throughout life.
- Published
- 2020
3. Generation of an apoptotic intracellular peptdide by g-secretase cleavage of Alzheimer’s amyliod b-protein precursor
- Author
-
Passer, B., Pellegrini, L., Russo, C., Siegel, Rm, Lenardo, Mj, Schettini, Gennaro, Bachmann, M., Tabaton, Massimo, and Dadamio, L.
- Published
- 2000
4. AMBRA1 controls the translation of immune-specific genes in T lymphocytes.
- Author
-
Gottlieb S, Shang W, Ye D, Kubo S, Jiang PD, Shafer S, Xu L, Zheng L, Park AY, Song J, Chan W, Zeng Z, He T, Schwarz B, Häupl B, Oellerich T, Lenardo MJ, and Yao Y
- Subjects
- Animals, Humans, Mice, CD28 Antigens metabolism, CD28 Antigens genetics, fas Receptor metabolism, fas Receptor genetics, Gene Expression Regulation, Lymphocyte Activation, Mechanistic Target of Rapamycin Complex 1 metabolism, Mechanistic Target of Rapamycin Complex 1 genetics, Phosphatidylinositol 3-Kinases metabolism, Protein Biosynthesis, Receptors, Antigen, T-Cell metabolism, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell immunology, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing genetics, Signal Transduction, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
T cell receptor (TCR) engagement causes a global cellular response that entrains signaling pathways, cell cycle regulation, and cell death. The molecular regulation of mRNA translation in these processes is poorly understood. Using a whole-genome CRISPR screen for regulators of CD95 (FAS/APO-1)-mediated T cell death, we identified AMBRA1, a protein previously studied for its roles in autophagy, E3 ubiquitin ligase activity, and cyclin regulation. T cells lacking AMBRA1 resisted FAS-mediated cell death by down-regulating FAS expression at the translational level. We show that AMBRA1 is a vital regulator of ribosome protein biosynthesis and ribosome loading on select mRNAs, whereby it plays a key role in balancing TCR signaling with cell cycle regulation pathways. We also found that AMBRA1 itself is translationally controlled by TCR stimulation via the CD28-PI3K-mTORC1-EIF4F pathway. Together, these findings shed light on the molecular control of translation after T cell activation and implicate AMBRA1 as a translational regulator governing TCR signaling, cell cycle progression, and T cell death., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
5. Germline mutations in a G protein identify signaling cross-talk in T cells.
- Author
-
Ham H, Jing H, Lamborn IT, Kober MM, Koval A, Berchiche YA, Anderson DE, Druey KM, Mandl JN, Isidor B, Ferreira CR, Freeman AF, Ganesan S, Karsak M, Mustillo PJ, Teo J, Zolkipli-Cunningham Z, Chatron N, Lecoquierre F, Oler AJ, Schmid JP, Kuhns DB, Xu X, Hauck F, Al-Herz W, Wagner M, Terhal PA, Muurinen M, Barlogis V, Cruz P, Danielson J, Stewart H, Loid P, Rading S, Keren B, Pfundt R, Zarember KA, Vill K, Potocki L, Olivier KN, Lesca G, Faivre L, Wong M, Puel A, Chou J, Tusseau M, Moutsopoulos NM, Matthews HF, Simons C, Taft RJ, Soldatos A, Masle-Farquhar E, Pittaluga S, Brink R, Fink DL, Kong HH, Kabat J, Kim WS, Bierhals T, Meguro K, Hsu AP, Gu J, Stoddard J, Banos-Pinero B, Slack M, Trivellin G, Mazel B, Soomann M, Li S, Watts VJ, Stratakis CA, Rodriguez-Quevedo MF, Bruel AL, Lipsanen-Nyman M, Saultier P, Jain R, Lehalle D, Torres D, Sullivan KE, Barbarot S, Neu A, Duffourd Y, Similuk M, McWalter K, Blanc P, Bézieau S, Jin T, Geha RS, Casanova JL, Makitie OM, Kubisch C, Edery P, Christodoulou J, Germain RN, Goodnow CC, Sakmar TP, Billadeau DD, Küry S, Katanaev VL, Zhang Y, Lenardo MJ, and Su HC
- Subjects
- Humans, Cell Movement genetics, Cell Proliferation, Immunity genetics, MAP Kinase Signaling System, Phosphatidylinositol 3-Kinases metabolism, Phosphatidylinositol 3-Kinases genetics, Proto-Oncogene Proteins c-akt metabolism, ras Proteins metabolism, ras Proteins genetics, Signal Transduction, Pedigree, Germ-Line Mutation, GTP-Binding Protein alpha Subunit, Gi2 genetics, ras GTPase-Activating Proteins genetics, Receptors, Antigen, T-Cell metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
Humans with monogenic inborn errors responsible for extreme disease phenotypes can reveal essential physiological pathways. We investigated germline mutations in GNAI2 , which encodes G
αi2 , a key component in heterotrimeric G protein signal transduction usually thought to regulate adenylyl cyclase-mediated cyclic adenosine monophosphate (cAMP) production. Patients with activating Gαi2 mutations had clinical presentations that included impaired immunity. Mutant Gαi2 impaired cell migration and augmented responses to T cell receptor (TCR) stimulation. We found that mutant Gαi2 influenced TCR signaling by sequestering the guanosine triphosphatase (GTPase)-activating protein RASA2, thereby promoting RAS activation and increasing downstream extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)-AKT S6 signaling to drive cellular growth and proliferation.- Published
- 2024
- Full Text
- View/download PDF
6. Pozelimab for CHAPLE disease: results from in-trial interviews and clinical outcome assessments.
- Author
-
Litcher-Kelly L, Ozen A, Ollis S, Feldman HB, Yaworsky A, Medrano P, Chongsrisawa V, Brackin T, Perlee L, Walker M, Pradeep S, Lenardo MJ, Harari OA, and Jalbert JJ
- Subjects
- Humans, Female, Male, Adolescent, Child, Preschool, Child, Young Adult, Adult, Outcome Assessment, Health Care, Antibodies, Monoclonal, Humanized therapeutic use
- Abstract
Background: CD55 deficiency with hyper-activation of complement, angiopathic thrombosis, and protein-losing enteropathy (CHAPLE) disease is ultra-rare (< 100 children or young adults worldwide) and potentially fatal. The study used mixed-methods approaches to assess how pozelimab impacts the signs and symptoms of CHAPLE disease from the patient perspective by combining within-trial interviews and clinical outcome assessments (COAs) (ClinicalTrials.gov, NCT04209634)., Methods: Interviews conducted with patients/caregivers at screening and week 24 assessed the signs and symptoms of CHAPLE disease, including those which were most bothersome, and evaluated the change. Patients/caregivers and clinicians completed the COAs; interview data contextualized the meaningfulness of change., Results: Ten patients (aged 3-19 years) were enrolled; caregivers contributed to nine interviews. Abdominal pain, diarrhea, facial and peripheral edema, nausea, and vomiting are the core signs and symptoms of CHAPLE disease (≥ 90% patients experienced pre-treatment); the most bothersome signs and symptoms were abdominal pain (n = 9) and facial edema (n = 1). All core signs and symptoms were reported as resolved at week 24 interviews. Severity on global assessments changed from "mild" to "very severe" at baseline to "no signs or symptoms" at week 24. Interview results were generally consistent with sign- or symptom-specific COA scores., Conclusions: Patients with CHAPLE disease treated with pozelimab for 24 weeks experienced complete resolution of core signs and symptoms. Mixed-methods approaches can contextualize the patient experience (how patients feel and function) in rare disease trials., Trial Registration: Clinicaltrials.gov, NCT04209634, registered December 24, 2019, https://classic., Clinicaltrials: gov/ct2/show/NCT04209634 ., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. The Canadian Collaborative Project on Genetic Susceptibility to Multiple Sclerosis cohort population structure and disease etiology.
- Author
-
Pagalilauan AM, Everest E, Rachimi S, Reich D, Waldman AD, Sadovnick AD, Vilariño-Guell C, and Lenardo MJ
- Abstract
Background: Previous genetic and epidemiological studies have examined subpopulations from the Canadian Collaborative Project on Genetic Susceptibility to Multiple Sclerosis (CCPGSMS) patient cohort, but an encompassing analysis of the study population has not yet been carried out., Objective: This study examines patterns of multiple sclerosis (MS) prevalence in 13,663 cohort members, including 4,821 patients with MS or suspected MS and 8,842 family members., Methods: We grouped participants into epidemiologic subgroups based on age of MS onset, clinical stage at diagnosis, symptom type at disease onset, sex, proband status, disability as measured by the EDSS, and ancestry based on reported ethnicity., Results: We observed a 2.7:1 MS prevalence ratio of women to men, though disease severity was greater for male patients. Variation in the age of disease onset between patients was only slightly associated with sex and strongly associated with disease type. Specific types of clinical symptoms at disease onset were associated with the prognosis. Regional residence did not correlate with disease onset, type, or severity., Conclusion: Population trends, as presented here, are not explained by environmental factors alone, highlighting the need for a comprehensive genetic analysis to understand disease variance across families.
- Published
- 2024
- Full Text
- View/download PDF
8. Author Correction: GIMAP5 deficiency reveals a mammalian ceramide-driven longevity assurance pathway.
- Author
-
Park AY, Leney-Greene M, Lynberg M, Gabrielski JQ, Xu X, Schwarz B, Zheng L, Balasubramaniyam A, Ham H, Chao B, Zhang Y, Matthews HF, Cui J, Yao Y, Kubo S, Chanchu JM, Morawski AR, Cook SA, Jiang P, Ravell JC, Cheng YH, George A, Faruqi A, Pagalilauan AM, Bergerson JRE, Ganesan S, Chauvin SD, Aluri J, Edwards-Hicks J, Bohrnsen E, Tippett C, Omar H, Xu L, Butcher GW, Pascall J, Karakoc-Aydiner E, Kiykim A, Maecker H, Tezcan İ, Esenboga S, Heredia RJ, Akata D, Tekin S, Kara A, Kuloglu Z, Unal E, Kendirli T, Dogu F, Karabiber E, Atkinson TP, Cochet C, Filhol O, Bosio CM, Davis MM, Lifton RP, Pearce EL, Daumke O, Aytekin C, Şahin GE, Aksu AÜ, Uzel G, Koneti Rao V, Sari S, Dalgıç B, Boztug K, Cagdas D, Haskologlu S, Ikinciogullari A, Schwefel D, Vilarinho S, Baris S, Ozen A, Su HC, and Lenardo MJ
- Published
- 2024
- Full Text
- View/download PDF
9. The prevalence and topography of spinal cord demyelination in multiple sclerosis: a retrospective study.
- Author
-
Waldman AD, Catania C, Pisa M, Jenkinson M, Lenardo MJ, and DeLuca GC
- Subjects
- Humans, Retrospective Studies, Prevalence, Spinal Cord pathology, Magnetic Resonance Imaging, Multiple Sclerosis pathology, Multiple Sclerosis, Chronic Progressive pathology
- Abstract
Spinal cord pathology is a major determinant of irreversible disability in progressive multiple sclerosis. The demyelinated lesion is a cardinal feature. The well-characterised anatomy of the spinal cord and new analytic approaches allows the systematic study of lesion topography and its extent of inflammatory activity unveiling new insights into disease pathogenesis. We studied cervical, thoracic, and lumbar spinal cord tissue from 119 pathologically confirmed multiple sclerosis cases. Immunohistochemistry was used to detect demyelination (PLP) and classify lesional inflammatory activity (CD68). Prevalence and distribution of demyelination, staged by lesion activity, was determined and topographical maps were created to identify patterns of lesion prevalence and distribution using mixed models and permutation-based voxelwise analysis. 460 lesions were observed throughout the spinal cord with 76.5% of cases demonstrating at least 1 lesion. The cervical level was preferentially affected by lesions. 58.3% of lesions were inflammatory with 87.9% of cases harbouring at least 1 inflammatory lesion. Topographically, lesions consistently affected the dorsal and lateral columns with relative sparing of subpial areas in a distribution mirroring the vascular network. The presence of spinal cord lesions and the proportion of active lesions related strongly with clinical disease milestones, including time from onset to wheelchair and onset to death. We demonstrate that spinal cord demyelination is common, highly inflammatory, has a predilection for the cervical level, and relates to clinical disability. The topography of lesions in the dorsal and lateral columns and relative sparing of subpial areas points to a role of the vasculature in lesion pathogenesis, suggesting short-range cell infiltration from the blood and signaling molecules circulating in the perivascular space incite lesion development. These findings challenge the notion that end-stage progressive multiple sclerosis is 'burnt out' and an outside-in lesional gradient predominates in the spinal cord. Taken together, this study provides support for long-term targeting of inflammatory demyelination in the spinal cord and nominates vascular dysfunction as a potential target for new therapeutic approaches to limit irreversible disability., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Evaluating the efficacy and safety of pozelimab in patients with CD55 deficiency with hyperactivation of complement, angiopathic thrombosis, and protein-losing enteropathy disease: an open-label phase 2 and 3 study.
- Author
-
Ozen A, Chongsrisawat V, Sefer AP, Kolukisa B, Jalbert JJ, Meagher KA, Brackin T, Feldman HB, Baris S, Karakoc-Aydiner E, Ergelen R, Fuss IJ, Moorman H, Suratannon N, Suphapeetiporn K, Perlee L, Harari OA, Yancopoulos GD, and Lenardo MJ
- Subjects
- Child, Humans, Antibodies, Monoclonal, Edema, Serum Albumin, Treatment Outcome, Historically Controlled Study, Male, Female, Protein-Losing Enteropathies drug therapy, Thrombosis
- Abstract
Background: CD55 deficiency with hyperactivation of complement, angiopathic thrombosis, and protein-losing enteropathy (CHAPLE) is an ultra-rare genetic disorder characterised by intestinal lymphatic damage, lymphangiectasia, and protein-losing enteropathy caused by overactivation of the complement system. We assessed the efficacy and safety of pozelimab, an antibody blocking complement component 5., Methods: This open-label, single-arm, historically controlled, multicentre phase 2 and 3 study evaluated ten patients with CHAPLE disease. This study was conducted at three hospitals in Thailand, Türkiye, and the USA. Patients aged 1 year or older with a clinical diagnosis of CHAPLE disease and a CD55 loss-of-function variant identified by genetic analysis and confirmed by flow cytometry or western blot of CD55 from peripheral blood cells were eligible for this study. Patients received a single intravenous loading dose of pozelimab 30 mg per kg of bodyweight, followed by a once-per-week subcutaneous dose over the treatment period based on bodyweight at a concentration of 200 mg/mL as either a single injection (<40 kg bodyweight) or two injections (≥40 kg bodyweight). The primary endpoint was proportion of patients with serum albumin normalisation with an improvement in active clinical outcomes and no worsening in inactive clinical outcomes (frequency of problematic abdominal pain, bowel movement frequency, facial oedema severity, and peripheral oedema severity) at week 24 compared with baseline, assessed in the full analysis set. This study is registered with ClinicalTrials.gov (NCT04209634) and is active but not recruiting., Findings: 11 patients were recruited between Jan 27, 2020, and May 12, 2021, ten of which were enrolled in the study and included in the analysis populations. The efficacy data corresponded to all patients completing the week 48 assessment and having at least 52 weeks of treatment exposure, and the safety data included an additional 90 days of follow-up and corresponded to all patients having at least 72 weeks of treatment. Patients were predominantly paediatric (with a median age of 8·5 years), and originated from Türkiye, Syria, Thailand, and Bolivia. Patients had markedly low weight-for-age and stature-for-age at baseline, and mean albumin at baseline was 2·2 g/dL, which was considerably less than the local laboratory reference range. After pozelimab treatment, all ten patients had serum albumin normalisation and improvement with no worsening in clinical outcomes. There was a complete inhibition of the total complement activity. Nine patients had adverse events; two were severe events, and one patient had an adverse event considered related to pozelimab., Interpretation: Pozelimab inhibits complement overactivation and resolves the clinical and laboratory manifestations of CHAPLE disease. Pozelimab is the only currently approved therapeutic drug for patients with this life-threatening, ultra-rare condition. In patients with protein-losing enteropathy where known causes have been excluded, testing for a CD55 deficiency should be contemplated. A diagnosis of CHAPLE disease should lead to early consideration of treatment with pozelimab., Funding: Regeneron Pharmaceuticals and the Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health., Competing Interests: Declaration of interests AO is a consultant and steering committee member for Regeneron Pharmaceuticals; received sample analysis support for a previous collaborative study (https://doi.org/10.1038/s41590-020-00830-z) from Regeneron Pharmaceuticals; and has a pending patent on component 5 inhibitor treatment in CHAPLE disease. VC, NS, and KS received support to conduct the study and received provision of the investigational product from Regeneron Pharmaceuticals. JJJ, TB, and LP are Regeneron Pharmaceuticals employees and stockholders. KAM is a Regeneron Pharmaceuticals employee and stockholder and has both pending and issued patents with Regeneron Pharmaceuticals. HBF is a consultant and advisory board member for Regeneron Pharmaceuticals. IJF is an associate on a cooperative research and development agreement between Merck Pharmaceuticals and the National Institutes for Health. OAH and GDY are Regeneron Pharmaceuticals employees and stockholders and have a pending patent on complement component 5 inhibitor treatment in CHAPLE disease. MJL received support for a federally approved cooperative research and development agreement to support the clinical trial and has a pending patent on complement component 5 inhibitor treatment in CHAPLE disease. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. GIMAP5 deficiency reveals a mammalian ceramide-driven longevity assurance pathway.
- Author
-
Park AY, Leney-Greene M, Lynberg M, Gabrielski JQ, Xu X, Schwarz B, Zheng L, Balasubramaniyam A, Ham H, Chao B, Zhang Y, Matthews HF, Cui J, Yao Y, Kubo S, Chanchu JM, Morawski AR, Cook SA, Jiang P, Ravell JC, Cheng YH, George A, Faruqi A, Pagalilauan AM, Bergerson JRE, Ganesan S, Chauvin SD, Aluri J, Edwards-Hicks J, Bohrnsen E, Tippett C, Omar H, Xu L, Butcher GW, Pascall J, Karakoc-Aydiner E, Kiykim A, Maecker H, Tezcan İ, Esenboga S, Heredia RJ, Akata D, Tekin S, Kara A, Kuloglu Z, Unal E, Kendirli T, Dogu F, Karabiber E, Atkinson TP, Cochet C, Filhol O, Bosio CM, Davis MM, Lifton RP, Pearce EL, Daumke O, Aytekin C, Şahin GE, Aksu AÜ, Uzel G, Koneti Rao V, Sari S, Dalgıç B, Boztug K, Cagdas D, Haskologlu S, Ikinciogullari A, Schwefel D, Vilarinho S, Baris S, Ozen A, Su HC, and Lenardo MJ
- Subjects
- Animals, Humans, Longevity genetics, Endothelial Cells metabolism, Mammals metabolism, GTP-Binding Proteins, Ceramides
- Abstract
Preserving cells in a functional, non-senescent state is a major goal for extending human healthspans. Model organisms reveal that longevity and senescence are genetically controlled, but how genes control longevity in different mammalian tissues is unknown. Here, we report a new human genetic disease that causes cell senescence, liver and immune dysfunction, and early mortality that results from deficiency of GIMAP5, an evolutionarily conserved GTPase selectively expressed in lymphocytes and endothelial cells. We show that GIMAP5 restricts the pathological accumulation of long-chain ceramides (CERs), thereby regulating longevity. GIMAP5 controls CER abundance by interacting with protein kinase CK2 (CK2), attenuating its ability to activate CER synthases. Inhibition of CK2 and CER synthase rescues GIMAP5-deficient T cells by preventing CER overaccumulation and cell deterioration. Thus, GIMAP5 controls longevity assurance pathways crucial for immune function and healthspan in mammals., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
12. Interim analysis: Open-label extension study of leniolisib for patients with APDS.
- Author
-
Rao VK, Kulm E, Šedivá A, Plebani A, Schuetz C, Shcherbina A, Dalm VA, Trizzino A, Zharankova Y, Webster S, Orpia A, Körholz J, Lougaris V, Rodina Y, Radford K, Bradt J, Relan A, Holland SM, Lenardo MJ, and Uzel G
- Subjects
- Humans, Female, Young Adult, Adult, Male, Class I Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases genetics, Quality of Life, Mutation, Immunologic Deficiency Syndromes genetics, Lymphadenopathy complications
- Abstract
Background: Activated phosphoinositide 3-kinase delta (PI3Kδ) syndrome (APDS; or p110δ-activating mutations causing senescent T cells, lymphadenopathy, and immunodeficiency) is an inborn error of immunity caused by PI3Kδ hyperactivity. Resultant immune deficiency and dysregulation lead to recurrent sinopulmonary infections, herpes viremia, autoimmunity, and lymphoproliferation., Objective: Leniolisib, a selective PI3Kδ inhibitor, demonstrated favorable impact on immune cell subsets and lymphoproliferation over placebo in patients with APDS over 12 weeks. Here, we report results from an interim analysis of an ongoing open-label, single-arm extension study., Methods: Patients with APDS aged 12 years or older who completed NCT02435173 or had previous exposure to PI3Kδ inhibitors were eligible. The primary end point was safety, assessed via investigator-reported adverse events (AEs) and clinical/laboratory evaluations. Secondary and exploratory end points included health-related quality of life, inflammatory markers, frequency of infections, and lymphoproliferation., Results: Between September 2016 and August 2021, 37 patients (median age, 20 years; 42.3% female) were enrolled. Of these 37 patients, 26, 9, and 2 patients had previously received leniolisib, placebo, or other PI3Kδ inhibitors, respectively. At the data cutoff date (December 13, 2021), median leniolisib exposure was 102 weeks. Overall, 32 patients (87%) experienced an AE. Most AEs were grades 1 to 3; none were grade 4. One patient with severe baseline comorbidities experienced a grade 5 AE, determined as unrelated to leniolisib treatment. While on leniolisib, patients had reduced annualized infection rates (P = .004), and reductions in immunoglobulin replacement therapy occurred in 10 of 27 patients. Other observations include reduced lymphadenopathy and splenomegaly, improved cytopenias, and normalized lymphocyte subsets., Conclusions: Leniolisib was well tolerated and maintained durable outcomes with up to 5 years of exposure in 37 patients with APDS., Clinicaltrials: gov identifier: NCT02859727., (Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
13. Precision medicine in monogenic inflammatory bowel disease: proposed mIBD REPORT standards.
- Author
-
Uhlig HH, Booth C, Cho J, Dubinsky M, Griffiths AM, Grimbacher B, Hambleton S, Huang Y, Jones K, Kammermeier J, Kanegane H, Koletzko S, Kotlarz D, Klein C, Lenardo MJ, Lo B, McGovern DPB, Özen A, de Ridder L, Ruemmele F, Shouval DS, Snapper SB, Travis SP, Turner D, Wilson DC, and Muise AM
- Subjects
- Humans, Prospective Studies, Retrospective Studies, Precision Medicine
- Abstract
Owing to advances in genomics that enable differentiation of molecular aetiologies, patients with monogenic inflammatory bowel disease (mIBD) potentially have access to genotype-guided precision medicine. In this Expert Recommendation, we review the therapeutic research landscape of mIBD, the reported response to therapies, the medication-related risks and systematic bias in reporting. The mIBD field is characterized by the absence of randomized controlled trials and is dominated by retrospective observational data based on case series and case reports. More than 25 off-label therapeutics (including small-molecule inhibitors and biologics) as well as cellular therapies (including haematopoietic stem cell transplantation and gene therapy) have been reported. Heterogeneous reporting of outcomes impedes the generation of robust therapeutic evidence as the basis for clinical decision making in mIBD. We discuss therapeutic goals in mIBD and recommend standardized reporting (mIBD REPORT (monogenic Inflammatory Bowel Disease Report Extended Phenotype and Outcome of Treatments) standards) to stratify patients according to a genetic diagnosis and phenotype, to assess treatment effects and to record safety signals. Implementation of these pragmatic standards should help clinicians to assess the therapy responses of individual patients in clinical practice and improve comparability between observational retrospective studies and controlled prospective trials, supporting future meta-analysis., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
14. Protein-Losing Enteropathy. Reply.
- Author
-
Ozen A and Lenardo MJ
- Subjects
- Humans, Protein-Losing Enteropathies etiology
- Published
- 2023
- Full Text
- View/download PDF
15. Protein-Losing Enteropathy.
- Author
-
Ozen A and Lenardo MJ
- Subjects
- Humans, Protein-Losing Enteropathies diagnosis, Protein-Losing Enteropathies etiology
- Published
- 2023
- Full Text
- View/download PDF
16. A randomized, placebo-controlled phase 3 trial of the PI3Kδ inhibitor leniolisib for activated PI3Kδ syndrome.
- Author
-
Rao VK, Webster S, Šedivá A, Plebani A, Schuetz C, Shcherbina A, Conlon N, Coulter T, Dalm VA, Trizzino A, Zharankova Y, Kulm E, Körholz J, Lougaris V, Rodina Y, Radford K, Bradt J, Kucher K, Relan A, Holland SM, Lenardo MJ, and Uzel G
- Subjects
- Humans, Class I Phosphatidylinositol 3-Kinases, Pyridines, Double-Blind Method, Phosphatidylinositol 3-Kinases, Pyrimidines
- Abstract
Activated phosphoinositide 3-kinase delta (PI3Kδ) syndrome (APDS) is an inborn error of immunity with clinical manifestations including infections, lymphoproliferation, autoimmunity, enteropathy, bronchiectasis, increased risk of lymphoma, and early mortality. Hyperactive PI3Kδ signaling causes APDS and is selectively targeted with leniolisib, an oral, small molecule inhibitor of PI3Kδ. Here, 31 patients with APDS aged ≥12 years were enrolled in a global, phase 3, triple-blinded trial and randomized 2:1 to receive 70 mg leniolisib or placebo twice daily for 12 weeks. Coprimary outcomes were differences from baseline in the index lymph node size and the percentage of naïve B cells in peripheral blood, assessed as proxies for immune dysregulation and deficiency. Both primary outcomes were met: the difference in the adjusted mean change (95% confidence interval [CI]) between leniolisib and placebo for lymph node size was -0.25 (-0.38, -0.12; P = .0006; N = 26) and for percentage of naïve B cells, was 37.30 (24.06, 50.54; P = .0002; N = 13). Leniolisib reduced spleen volume compared with placebo (adjusted mean difference in 3-dimensional volume [cm3], -186; 95% CI, -297 to -76.2; P = .0020) and improved key immune cell subsets. Fewer patients receiving leniolisib reported study treatment-related adverse events (AEs; mostly grades 1-2) than those receiving placebo (23.8% vs 30.0%). Overall, leniolisib was well tolerated and significant improvement over placebo was notable in the coprimary endpoints, reducing lymphadenopathy and increasing the percentage of naïve B cells, reflecting a favorable impact on the immune dysregulation and deficiency seen in patients with APDS. This trial was registered at www.clinicaltrials.gov as #NCT02435173., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Death receptor 5 rises to the occasion.
- Author
-
Zheng L, Yao Y, and Lenardo MJ
- Subjects
- Receptors, TNF-Related Apoptosis-Inducing Ligand
- Published
- 2023
- Full Text
- View/download PDF
18. Mechanisms of antigen-induced reversal of CNS inflammation in experimental demyelinating disease.
- Author
-
Li J, Lu L, Binder K, Xiong J, Ye L, Cheng YH, Majri-Morrison S, Lu W, Lee JW, Zhang Z, Wu YZ, Zheng L, and Lenardo MJ
- Subjects
- Animals, Mice, Inflammation, Apoptosis, B-Lymphocytes, Encephalitis, Encephalomyelitis, Autoimmune, Experimental, Drug-Related Side Effects and Adverse Reactions
- Abstract
Autoimmune central nervous system (CNS) demyelinating diseases are a major public health burden and poorly controlled by current immunosuppressants. More precise immunotherapies with higher efficacy and fewer side effects are sought. We investigated the effectiveness and mechanism of an injectable myelin-based antigenic polyprotein MMPt (myelin oligodendrocyte glycoprotein, myelin basic protein and proteolipid protein, truncated). We find that it suppresses mouse experimental autoimmune encephalomyelitis without major side effects. MMPt induces rapid apoptosis of the encephalitogenic T cells and suppresses inflammation in the affected CNS. Intravital microscopy shows that MMPt is taken up by perivascular F4/80
+ cells but not conventional antigen-presenting dendritic cells, B cells, or microglia. MMPt-stimulated F4/80+ cells induce reactive T cell immobilization and apoptosis in situ, resulting in reduced infiltration of inflammatory cells and chemokine production. Our study reveals alternative mechanisms that explain how cognate antigen suppresses CNS inflammation and may be applicable for effectively and safely treating demyelinating diseases.- Published
- 2023
- Full Text
- View/download PDF
19. HEM1 Actin Immunodysregulatory Disorder: Genotypes, Phenotypes, and Future Directions.
- Author
-
Cook S, Lenardo MJ, and Freeman AF
- Subjects
- Humans, Autoimmunity genetics, Phenotype, Genotype, Membrane Proteins genetics, Actins genetics, Actins metabolism, Phagocytosis genetics
- Abstract
Cells of the innate and adaptive immune systems depend on proper actin dynamics to control cell behavior for effective immune responses. Dysregulated actin networks are known to play a pathogenic role in an increasing number of inborn errors of immunity. The WAVE regulatory complex (WRC) mediates branched actin polymerization, a process required for key cellular functions including migration, phagocytosis, vesicular transport, and immune synapse formation. Recent reports of pathogenic variants in NCKAP1L, a hematopoietically restricted gene encoding the HEM1 protein component of the WRC, defined a novel disease involving recurrent bacterial and viral infections, autoimmunity, and excessive inflammation (OMIM 141180). This review summarizes the diverse clinical presentations and immunological phenotypes observed in HEM1-deficient patients. In addition, we integrate the pathophysiological mechanisms described in current literature and highlight the outstanding questions for diagnosis and management of the HEM1 actin immunodysregulatory disorder., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2022
- Full Text
- View/download PDF
20. ERO1 alpha deficiency impairs angiogenesis by increasing N-glycosylation of a proangiogenic VEGFA.
- Author
-
Varone E, Chernorudskiy A, Cherubini A, Cattaneo A, Bachi A, Fumagalli S, Erol G, Gobbi M, Lenardo MJ, Borgese N, and Zito E
- Subjects
- Disulfides metabolism, Glycosylation, Humans, Lectins metabolism, Neovascularization, Pathologic genetics, Oxidation-Reduction, Oxidoreductases metabolism, Protein Folding, Thioredoxins metabolism, Membrane Glycoproteins metabolism, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor A metabolism
- Abstract
N-glycosylation and disulfide bond formation are two essential steps in protein folding that occur in the endoplasmic reticulum (ER) and reciprocally influence each other. Here, to analyze crosstalk between N-glycosylation and oxidation, we investigated how the protein disulfide oxidase ERO1-alpha affects glycosylation of the angiogenic VEGF
121 , a key regulator of vascular homeostasis. ERO1 deficiency, while retarding disulfide bond formation in VEGF121 , increased utilization of its single N-glycosylation sequon, which lies close to an intra-polypeptide disulfide bridge, and concomitantly slowed its secretion. Unbiased mass-spectrometric analysis revealed interactions between VEGF121 and N-glycosylation pathway proteins in ERO1-knockout (KO), but not wild-type cells. Notably, MAGT1, a thioredoxin-containing component of the post-translational oligosaccharyltransferase complex, was a major hit exclusive to ERO1-deficient cells. Thus, both a reduced rate of formation of disulfide bridges, and the increased trapping potential of MAGT1 may increase N-glycosylation of VEGF121 . Extending our investigation to tissues, we observed altered lectin staining of ERO1 KO breast tumor xenografts, implicating ERO1 as a physiologic regulator of protein N-glycosylation. Our study, highlighting the effect of ERO1 loss on N-glycosylation of proteins, is particularly relevant not only to angiogenesis but also to other cancer patho-mechanisms in light of recent findings suggesting a close causal link between alterations in protein glycosylation and cancer development., Competing Interests: Declaration of competing interest The authors declare no conflicting interests., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
21. Early B cell factor 4 modulates FAS-mediated apoptosis and promotes cytotoxic function in human immune cells.
- Author
-
Kubo S, Kataria R, Yao Y, Gabrielski JQ, Zheng L, Markowitz TE, Chan W, Song J, Boddapati AK, Saeki K, Häupl B, Park AY, Cheng YH, Cui J, Oellerich T, and Lenardo MJ
- Subjects
- Animals, Chromatin metabolism, Granzymes genetics, Humans, Mice, Perforin metabolism, Apoptosis physiology, CD8-Positive T-Lymphocytes, Cytotoxicity, Immunologic genetics, Fas Ligand Protein metabolism, T-Lymphocytes, Cytotoxic, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Apoptosis is a genetically regulated program of cell death that plays a key role in immune disease processes. We identified EBF4, a little-studied member of the early B cell factor (EBF) family of transcription factors, in a whole-genome CRISPR screen for regulators of Fas/APO-1/CD95-mediated T cell death. Loss of EBF4 increases the half-life of the c-FLIP protein, and its presence in the Fas signaling complex impairs caspase-8 cleavage and apoptosis. Transcriptome analysis revealed that EBF4 regulates molecules such as TBX21, EOMES, granzyme, and perforin that are important for human natural killer (NK) and CD8
+ T cell functions. Proximity-dependent biotin identification (Bio-ID) mass spectrometry analyses showed EBF4 binding to STAT3, STAT5, and MAP kinase 3 and a strong pathway relationship to interleukin-2 regulated genes, which are known to govern cytotoxicity pathways. Chromatin immunoprecipitation and DNA sequencing analysis defined a canonical EBF4 binding motif, 5'-CCCNNGG/AG-3', closely related to the EBF1 binding site; using a luciferase-based reporter, we found a dose-dependent transcriptional response of this motif to EBF4. We also conducted assay for transposase-accessible chromatin sequencing in EBF4-overexpressing cells and found increased chromatin accessibility upstream of granzyme and perforin and in topologically associated domains in human lymphocytes. Finally, we discovered that the EBF4 has basal expression in human but not mouse NK cells and CD8+ T cells and vanishes following activating stimulation. Together, our data reveal key features of a previously unknown transcriptional regulator of human cytotoxic immune function.- Published
- 2022
- Full Text
- View/download PDF
22. GIMAP6 regulates autophagy, immune competence, and inflammation in mice and humans.
- Author
-
Yao Y, Du Jiang P, Chao BN, Cagdas D, Kubo S, Balasubramaniyam A, Zhang Y, Shadur B, NaserEddin A, Folio LR, Schwarz B, Bohrnsen E, Zheng L, Lynberg M, Gottlieb S, Leney-Greene MA, Park AY, Tezcan I, Akdogan A, Gocmen R, Onder S, Rosenberg A, Soilleux EJ, Johnson E, Jackson PK, Demeter J, Chauvin SD, Paul F, Selbach M, Bulut H, Clatworthy MR, Tuong ZK, Zhang H, Stewart BJ, Bosio CM, Stepensky P, Clare S, Ganesan S, Pascall JC, Daumke O, Butcher GW, McMichael AJ, Simon AK, and Lenardo MJ
- Subjects
- Animals, Autophagy, Endothelial Cells metabolism, Humans, Inflammation, Mice, GTP Phosphohydrolases genetics, GTP Phosphohydrolases metabolism, Immunologic Deficiency Syndromes
- Abstract
Inborn errors of immunity (IEIs) unveil regulatory pathways of human immunity. We describe a new IEI caused by mutations in the GTPase of the immune-associated protein 6 (GIMAP6) gene in patients with infections, lymphoproliferation, autoimmunity, and multiorgan vasculitis. Patients and Gimap6-/- mice show defects in autophagy, redox regulation, and polyunsaturated fatty acid (PUFA)-containing lipids. We find that GIMAP6 complexes with GABARAPL2 and GIMAP7 to regulate GTPase activity. Also, GIMAP6 is induced by IFN-γ and plays a critical role in antibacterial immunity. Finally, we observed that Gimap6-/- mice died prematurely from microangiopathic glomerulosclerosis most likely due to GIMAP6 deficiency in kidney endothelial cells., (This is a work of the U.S. Government and is not subject to copyright protection in the United States. Foreign copyrights may apply.)
- Published
- 2022
- Full Text
- View/download PDF
23. Mucus sialylation determines intestinal host-commensal homeostasis.
- Author
-
Yao Y, Kim G, Shafer S, Chen Z, Kubo S, Ji Y, Luo J, Yang W, Perner SP, Kanellopoulou C, Park AY, Jiang P, Li J, Baris S, Aydiner EK, Ertem D, Mulder DJ, Warner N, Griffiths AM, Topf-Olivestone C, Kori M, Werner L, Ouahed J, Field M, Liu C, Schwarz B, Bosio CM, Ganesan S, Song J, Urlaub H, Oellerich T, Malaker SA, Zheng L, Bertozzi CR, Zhang Y, Matthews H, Montgomery W, Shih HY, Jiang J, Jones M, Baras A, Shuldiner A, Gonzaga-Jauregui C, Snapper SB, Muise AM, Shouval DS, Ozen A, Pan KT, Wu C, and Lenardo MJ
- Subjects
- Animals, Homeostasis, Humans, Intestinal Mucosa metabolism, Intestinal Mucosa microbiology, Mice, Mucus metabolism, Sialyltransferases metabolism, Symbiosis, Gastrointestinal Microbiome, Inflammatory Bowel Diseases genetics, Inflammatory Bowel Diseases metabolism, Sialyltransferases genetics
- Abstract
Intestinal mucus forms the first line of defense against bacterial invasion while providing nutrition to support microbial symbiosis. How the host controls mucus barrier integrity and commensalism is unclear. We show that terminal sialylation of glycans on intestinal mucus by ST6GALNAC1 (ST6), the dominant sialyltransferase specifically expressed in goblet cells and induced by microbial pathogen-associated molecular patterns, is essential for mucus integrity and protecting against excessive bacterial proteolytic degradation. Glycoproteomic profiling and biochemical analysis of ST6 mutations identified in patients show that decreased sialylation causes defective mucus proteins and congenital inflammatory bowel disease (IBD). Mice harboring a patient ST6 mutation have compromised mucus barriers, dysbiosis, and susceptibility to intestinal inflammation. Based on our understanding of the ST6 regulatory network, we show that treatment with sialylated mucin or a Foxo3 inhibitor can ameliorate IBD., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
24. A Double-Blind, Placebo-Controlled, Crossover Study of Magnesium Supplementation in Patients with XMEN Disease.
- Author
-
Chauvin SD, Price S, Zou J, Hunsberger S, Brofferio A, Matthews H, Similuk M, Rosenzweig SD, Su HC, Cohen JI, Lenardo MJ, and Ravell JC
- Subjects
- CD8-Positive T-Lymphocytes, Cross-Over Studies, Dietary Supplements, Herpesvirus 4, Human physiology, Humans, Magnesium metabolism, Magnesium therapeutic use, Cation Transport Proteins genetics, Epstein-Barr Virus Infections complications, Epstein-Barr Virus Infections drug therapy, Epstein-Barr Virus Infections genetics, Neoplasms genetics, X-Linked Combined Immunodeficiency Diseases genetics
- Abstract
X-linked MAGT1 deficiency with increased susceptibility to Epstein-Barr virus (EBV) infection and N-linked glycosylation defect (XMEN) disease is an inborn error of immunity caused by loss-of-function mutations in the magnesium transporter 1 (MAGT1) gene. The original studies of XMEN patients focused on impaired magnesium regulation, leading to decreased EBV-cytotoxicity and the loss of surface expression of the activating receptor "natural killer group 2D" (NKG2D) on CD8
+ T cells and NK cells. In vitro studies showed that supraphysiological supplementation of magnesium rescued these defects. Observational studies in 2 patients suggested oral magnesium supplementation could decrease EBV viremia. Hence, we performed a randomized, double-blind, placebo-controlled, crossover study in 2 parts. In part 1, patients received either oral magnesium L-threonate (MLT) or placebo for 12 weeks followed by 12 weeks of the other treatment. Part 2 began with 3 days of high-dose intravenous (IV) magnesium sulfate (MgSO4 ) followed by open-label MLT for 24 weeks. One EBV-infected and 3 EBV-naïve patients completed part 1. One EBV-naïve patient was removed from part 2 of the study due to asymptomatic elevation of liver enzymes during IV MgSO4 . No change in EBV or NKG2D status was observed. In vitro magnesium supplementation experiments in cells from 14 XMEN patients failed to significantly rescue NKG2D expression and the clinical trial was stopped. Although small, this study indicates magnesium supplementation is unlikely to be an effective therapeutic option in XMEN disease., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2022
- Full Text
- View/download PDF
25. Congenital iRHOM2 deficiency causes ADAM17 dysfunction and environmentally directed immunodysregulatory disease.
- Author
-
Kubo S, Fritz JM, Raquer-McKay HM, Kataria R, Vujkovic-Cvijin I, Al-Shaibi A, Yao Y, Zheng L, Zou J, Waldman AD, Jing X, Farley TK, Park AY, Oler AJ, Charles AK, Makhlouf M, AbouMoussa EH, Hasnah R, Saraiva LR, Ganesan S, Al-Subaiey AA, Matthews H, Flano E, Lee HH, Freeman AF, Sefer AP, Sayar E, Çakır E, Karakoc-Aydiner E, Baris S, Belkaid Y, Ozen A, Lo B, and Lenardo MJ
- Subjects
- A549 Cells, Animals, Child, Child, Preschool, Citrobacter rodentium pathogenicity, Colitis genetics, Cytokines genetics, Enterobacteriaceae Infections genetics, Female, HEK293 Cells, Humans, Infant, Newborn, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Mutation genetics, Pseudomonas Infections genetics, Pseudomonas aeruginosa pathogenicity, Signal Transduction genetics, ADAM17 Protein genetics, Carrier Proteins genetics, Primary Immunodeficiency Diseases genetics
- Abstract
We report a pleiotropic disease due to loss-of-function mutations in RHBDF2, the gene encoding iRHOM2, in two kindreds with recurrent infections in different organs. One patient had recurrent pneumonia but no colon involvement, another had recurrent infectious hemorrhagic colitis but no lung involvement and the other two experienced recurrent respiratory infections. Loss of iRHOM2, a rhomboid superfamily member that regulates the ADAM17 metalloproteinase, caused defective ADAM17-dependent cleavage and release of cytokines, including tumor-necrosis factor and amphiregulin. To understand the diverse clinical phenotypes, we challenged Rhbdf2
-/- mice with Pseudomonas aeruginosa by nasal gavage and observed more severe pneumonia, whereas infection with Citrobacter rodentium caused worse inflammatory colitis than in wild-type mice. The fecal microbiota in the colitis patient had characteristic oral species that can predispose to colitis. Thus, a human immunodeficiency arising from iRHOM2 deficiency causes divergent disease phenotypes that can involve the local microbial environment., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2022
- Full Text
- View/download PDF
26. CRISPR-targeted MAGT1 insertion restores XMEN patient hematopoietic stem cells and lymphocytes.
- Author
-
Brault J, Liu T, Bello E, Liu S, Sweeney CL, Meis RJ, Koontz S, Corsino C, Choi U, Vayssiere G, Bosticardo M, Dowdell K, Lazzarotto CR, Clark AB, Notarangelo LD, Ravell JC, Lenardo MJ, Kleinstiver BP, Tsai SQ, Wu X, Dahl GA, Malech HL, and De Ravin SS
- Subjects
- Animals, CRISPR-Cas Systems, Cation Transport Proteins deficiency, Cells, Cultured, Female, Genetic Therapy, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells pathology, Humans, Lymphocytes pathology, Male, Mice, Inbred NOD, X-Linked Combined Immunodeficiency Diseases pathology, X-Linked Combined Immunodeficiency Diseases therapy, Mice, Cation Transport Proteins genetics, Gene Editing methods, Hematopoietic Stem Cells metabolism, Lymphocytes metabolism, X-Linked Combined Immunodeficiency Diseases genetics
- Abstract
XMEN disease, defined as "X-linked MAGT1 deficiency with increased susceptibility to Epstein-Barr virus infection and N-linked glycosylation defect," is a recently described primary immunodeficiency marked by defective T cells and natural killer (NK) cells. Unfortunately, a potentially curative hematopoietic stem cell transplantation is associated with high mortality rates. We sought to develop an ex vivo targeted gene therapy approach for patients with XMEN using a CRISPR/Cas9 adeno-associated vector (AAV) to insert a therapeutic MAGT1 gene at the constitutive locus under the regulation of the endogenous promoter. Clinical translation of CRISPR/Cas9 AAV-targeted gene editing (GE) is hampered by low engraftable gene-edited hematopoietic stem and progenitor cells (HSPCs). Here, we optimized GE conditions by transient enhancement of homology-directed repair while suppressing AAV-associated DNA damage response to achieve highly efficient (>60%) genetic correction in engrafting XMEN HSPCs in transplanted mice. Restored MAGT1 glycosylation function in human NK and CD8+ T cells restored NK group 2 member D (NKG2D) expression and function in XMEN lymphocytes for potential treatment of infections, and it corrected HSPCs for long-term gene therapy, thus offering 2 efficient therapeutic options for XMEN poised for clinical translation., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
27. NF-κB Rel subunit exchange on a physiological timescale.
- Author
-
Biancalana M, Natan E, Lenardo MJ, and Fersht AR
- Subjects
- Binding Sites, Cloning, Molecular, DNA genetics, DNA metabolism, Escherichia coli genetics, Escherichia coli metabolism, Gene Expression, Genetic Vectors chemistry, Genetic Vectors metabolism, Humans, Kinetics, Models, Molecular, NF-kappa B p50 Subunit genetics, NF-kappa B p50 Subunit metabolism, Oligodeoxyribonucleotides metabolism, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Protein Multimerization, Protein Subunits genetics, Protein Subunits metabolism, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Thermodynamics, Transcription Factor RelA genetics, Transcription Factor RelA metabolism, DNA chemistry, NF-kappa B p50 Subunit chemistry, Oligodeoxyribonucleotides chemistry, Protein Subunits chemistry, Transcription Factor RelA chemistry
- Abstract
The Rel proteins of the NF-κB complex comprise one of the most investigated transcription factor families, forming a variety of hetero- or homodimers. Nevertheless, very little is known about the fundamental kinetics of NF-κB complex assembly, or the inter-conversion potential of dimerised Rel subunits. Here, we examined an unexplored aspect of NF-κB dynamics, focusing on the dissociation and reassociation of the canonical p50 and p65 Rel subunits and their ability to form new hetero- or homodimers. We employed a soluble expression system to enable the facile production of NF-κB Rel subunits, and verified these proteins display canonical NF-κB nucleic acid binding properties. Using a combination of biophysical techniques, we demonstrated that, at physiological temperatures, homodimeric Rel complexes routinely exchange subunits with a half-life of less than 10 min. In contrast, we found a dramatic preference for the formation of the p50/p65 heterodimer, which demonstrated a kinetic stability of at least an order of magnitude greater than either homodimer. These results suggest that specific DNA targets of either the p50 or p65 homodimers can only be targeted when these subunits are expressed exclusively, or with the intervention of additional post-translational modifications. Together, this work implies a new model of how cells can modulate NF-κB activity by fine-tuning the relative proportions of the p50 and p65 proteins, as well as their time of expression. This work thus provides a new quantitative interpretation of Rel dimer distribution in the cell, particularly for those who are developing mathematical models of NF-κB activity., (© 2021 The Protein Society.)
- Published
- 2021
- Full Text
- View/download PDF
28. GIMAP5 maintains liver endothelial cell homeostasis and prevents portal hypertension.
- Author
-
Drzewiecki K, Choi J, Brancale J, Leney-Greene MA, Sari S, Dalgiç B, Ünlüsoy Aksu A, Evirgen Şahin G, Ozen A, Baris S, Karakoc-Aydiner E, Jain D, Kleiner D, Schmalz M, Radhakrishnan K, Zhang J, Hoebe K, Su HC, Pereira JP, Lenardo MJ, Lifton RP, and Vilarinho S
- Subjects
- Adolescent, Adult, Animals, Female, Hepatocytes metabolism, Humans, Liver Cirrhosis metabolism, Male, Mice, Young Adult, Endothelial Cells metabolism, GTP-Binding Proteins metabolism, Homeostasis physiology, Hypertension, Portal metabolism, Liver metabolism
- Abstract
Portal hypertension is a major contributor to decompensation and death from liver disease, a global health problem. Here, we demonstrate homozygous damaging mutations in GIMAP5, a small organellar GTPase, in four families with unexplained portal hypertension. We show that GIMAP5 is expressed in hepatic endothelial cells and that its loss in both humans and mice results in capillarization of liver sinusoidal endothelial cells (LSECs); this effect is also seen when GIMAP5 is selectively deleted in endothelial cells. Single-cell RNA-sequencing analysis in a GIMAP5-deficient mouse model reveals replacement of LSECs with capillarized endothelial cells, a reduction of macrovascular hepatic endothelial cells, and places GIMAP5 upstream of GATA4, a transcription factor required for LSEC specification. Thus, GIMAP5 is a critical regulator of liver endothelial cell homeostasis and, when absent, produces portal hypertension. These findings provide new insight into the pathogenesis of portal hypertension, a major contributor to morbidity and mortality from liver disease., Competing Interests: Disclosures: The authors declare no competing interests exist., (© 2021 Drzewiecki et al.)
- Published
- 2021
- Full Text
- View/download PDF
29. Ulrich Siebenlist (1951-2020).
- Author
-
Murphy PM and Lenardo MJ
- Subjects
- Gene Expression Regulation, History, 20th Century, History, 21st Century, Humans, Immunoglobulin D genetics, Male, United States, Allergy and Immunology history, NF-kappa B genetics, Signal Transduction immunology
- Published
- 2021
- Full Text
- View/download PDF
30. Homozygous IL37 mutation associated with infantile inflammatory bowel disease.
- Author
-
Zhang ZZ, Zhang Y, He T, Sweeney CL, Baris S, Karakoc-Aydiner E, Yao Y, Ertem D, Matthews HF, Gonzaga-Jauregui C, Malech HL, Su HC, Ozen A, Smith KGC, and Lenardo MJ
- Subjects
- Child, Preschool, Female, Humans, Induced Pluripotent Stem Cells immunology, Interleukin-18 genetics, Interleukin-18 immunology, Interleukin-1beta genetics, Interleukin-1beta immunology, Macrophage Activation genetics, Male, Gene Expression Regulation immunology, Inflammatory Bowel Diseases genetics, Inflammatory Bowel Diseases immunology, Interleukin-1 genetics, Interleukin-1 immunology, Loss of Function Mutation, Macrophage Activation immunology, Macrophages immunology
- Abstract
Interleukin (IL)-37, an antiinflammatory IL-1 family cytokine, is a key suppressor of innate immunity. IL-37 signaling requires the heterodimeric IL-18R1 and IL-1R8 receptor, which is abundantly expressed in the gastrointestinal tract. Here we report a 4-mo-old male from a consanguineous family with a homozygous loss-of-function IL37 mutation. The patient presented with persistent diarrhea and was found to have infantile inflammatory bowel disease (I-IBD). Patient cells showed increased intracellular IL-37 expression and increased proinflammatory cytokine production. In cell lines, mutant IL-37 was not stably expressed or properly secreted and was thus unable to functionally suppress proinflammatory cytokine expression. Furthermore, induced pluripotent stem cell-derived macrophages from the patient revealed an activated macrophage phenotype, which is more prone to lipopolysaccharide and IL-1β stimulation, resulting in hyperinflammatory tumor necrosis factor production. Insights from this patient will not only shed light on monogenic contributions of I-IBD but may also reveal the significance of the IL-18 and IL-37 axis in colonic homeostasis., Competing Interests: Competing interest statement: C.G.-J. is a full-time employee of the Regeneron Genetics Center and receives stock options as part of compensation., (Copyright © 2021 the Author(s). Published by PNAS.)
- Published
- 2021
- Full Text
- View/download PDF
31. MAGT1 messenger RNA-corrected autologous T and natural killer cells for potential cell therapy in X-linked immunodeficiency with magnesium defect, Epstein-Barr virus infection and neoplasia disease.
- Author
-
Brault J, Meis RJ, Li L, Bello E, Liu T, Sweeney CL, Koontz SM, Dowdell K, Theobald N, Lee J, Allen C, Clark AB, Ravell JC, Lenardo MJ, Dahl GA, Malech HL, and De Ravin SS
- Subjects
- Cell- and Tissue-Based Therapy, Herpesvirus 4, Human genetics, Humans, Killer Cells, Natural metabolism, Magnesium metabolism, RNA, Messenger genetics, Cation Transport Proteins, Epstein-Barr Virus Infections, Neoplasms
- Abstract
Background Aim: X-linked MAGT1 deficiency with increased susceptibility to EBV-infection and N-linked glycosylation defect' (XMEN) disease is caused by mutations in the magnesium transporter 1 (MAGT1) gene. Loss of MAGT1 function results in a glycosylation defect that abrogates expression of key immune proteins such as the NKG2D receptor on CD8
+ T and NK cells, which is critical for the recognition and killing of virus-infected and transformed cells, a biomarker for MAGT1 function. Patients with XMEN disease frequently have increased susceptibility to EBV infections and EBV-associated B cell malignancies, for which no specific treatment options are currently available. Experimental transfer of donor EBV-specific cytotoxic T cells may be beneficial but carries the risks of eliciting alloimmune responses. An approach for cell therapy to address viral infections and associated complications that avoids the risks of alloimmunity is needed., Methods: Here the authors assess the feasibility and efficiency of correcting autologous lymphocytes from XMEN patients by MAGT1 mRNA electroporation (EP) that avoids genomic integration and can be scaled for clinical application., Results and Conclusions: Restoration of NKG2D expression was demonstrated in XMEN patient lymphocytes after MAGT1 mRNA electroporation that reach healthy donor levels in CD8+ T and NK cells at 1-2 days after EP. NKG2D expression persisted at ∼50% for 2 weeks after EP. Functionally, mRNA-correction of XMEN NK cells rescued cytotoxic activity also to healthy donor NK cell level. The restored NKG2D receptor expression and function were unaffected by cryopreservation, which will make feasible repeat infusions of MAGT1 mRNA-corrected autologous XMEN CD8+ T and NK cells for potential short term therapy for XMEN patients without the risks of alloimmunization., (Published by Elsevier Inc.)- Published
- 2021
- Full Text
- View/download PDF
32. Broadly effective metabolic and immune recovery with C5 inhibition in CHAPLE disease.
- Author
-
Ozen A, Kasap N, Vujkovic-Cvijin I, Apps R, Cheung F, Karakoc-Aydiner E, Akkelle B, Sari S, Tutar E, Ozcay F, Uygun DK, Islek A, Akgun G, Selcuk M, Sezer OB, Zhang Y, Kutluk G, Topal E, Sayar E, Celikel C, Houwen RHJ, Bingol A, Ogulur I, Eltan SB, Snow AL, Lake C, Fantoni G, Alba C, Sellers B, Chauvin SD, Dalgard CL, Harari O, Ni YG, Wang MD, Devalaraja-Narashimha K, Subramanian P, Ergelen R, Artan R, Guner SN, Dalgic B, Tsang J, Belkaid Y, Ertem D, Baris S, and Lenardo MJ
- Subjects
- Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized pharmacokinetics, Biomarkers blood, CD55 Antigens deficiency, CD55 Antigens genetics, Complement C5 metabolism, Complement Inactivating Agents adverse effects, Complement Inactivating Agents pharmacokinetics, Genetic Predisposition to Disease, Humans, Hypoproteinemia genetics, Hypoproteinemia immunology, Hypoproteinemia metabolism, Mutation, Phenotype, Protein-Losing Enteropathies genetics, Protein-Losing Enteropathies immunology, Protein-Losing Enteropathies metabolism, Treatment Outcome, Antibodies, Monoclonal, Humanized therapeutic use, Complement Activation drug effects, Complement C5 antagonists & inhibitors, Complement Inactivating Agents therapeutic use, Energy Metabolism drug effects, Hypoproteinemia drug therapy, Immunity, Innate drug effects, Protein-Losing Enteropathies drug therapy
- Abstract
Complement hyperactivation, angiopathic thrombosis and protein-losing enteropathy (CHAPLE disease) is a lethal disease caused by genetic loss of the complement regulatory protein CD55, leading to overactivation of complement and innate immunity together with immunodeficiency due to immunoglobulin wasting in the intestine. We report in vivo human data accumulated using the complement C5 inhibitor eculizumab for the medical treatment of patients with CHAPLE disease. We observed cessation of gastrointestinal pathology together with restoration of normal immunity and metabolism. We found that patients rapidly renormalized immunoglobulin concentrations and other serum proteins as revealed by aptamer profiling, re-established a healthy gut microbiome, discontinued immunoglobulin replacement and other treatments and exhibited catch-up growth. Thus, we show that blockade of C5 by eculizumab effectively re-establishes regulation of the innate immune complement system to substantially reduce the pathophysiological manifestations of CD55 deficiency in humans.
- Published
- 2021
- Full Text
- View/download PDF
33. Extended clinical and immunological phenotype and transplant outcome in CD27 and CD70 deficiency.
- Author
-
Ghosh S, Köstel Bal S, Edwards ESJ, Pillay B, Jiménez Heredia R, Erol Cipe F, Rao G, Salzer E, Zoghi S, Abolhassani H, Momen T, Gostick E, Price DA, Zhang Y, Oler AJ, Gonzaga-Jauregui C, Erman B, Metin A, Ilhan I, Haskologlu S, Islamoglu C, Baskin K, Ceylaner S, Yilmaz E, Unal E, Karakukcu M, Berghuis D, Cole T, Gupta AK, Hauck F, Kogler H, Hoepelman AIM, Baris S, Karakoc-Aydiner E, Ozen A, Kager L, Holzinger D, Paulussen M, Krüger R, Meisel R, Oommen PT, Morris E, Neven B, Worth A, van Montfrans J, Fraaij PLA, Choo S, Dogu F, Davies EG, Burns S, Dückers G, Becker RP, von Bernuth H, Latour S, Faraci M, Gattorno M, Su HC, Pan-Hammarström Q, Hammarström L, Lenardo MJ, Ma CS, Niehues T, Aghamohammadi A, Rezaei N, Ikinciogullari A, Tangye SG, Lankester AC, and Boztug K
- Subjects
- Adolescent, Adult, Allografts, Child, Child, Preschool, Disease-Free Survival, Female, Humans, Infant, Male, Retrospective Studies, Survival Rate, CD27 Ligand deficiency, Genetic Diseases, Inborn genetics, Genetic Diseases, Inborn immunology, Genetic Diseases, Inborn mortality, Genetic Diseases, Inborn therapy, Hematopoietic Stem Cell Transplantation, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Immunologic Deficiency Syndromes mortality, Immunologic Deficiency Syndromes therapy, Tumor Necrosis Factor Receptor Superfamily, Member 7 deficiency
- Abstract
Biallelic mutations in the genes encoding CD27 or its ligand CD70 underlie inborn errors of immunity (IEIs) characterized predominantly by Epstein-Barr virus (EBV)-associated immune dysregulation, such as chronic viremia, severe infectious mononucleosis, hemophagocytic lymphohistiocytosis (HLH), lymphoproliferation, and malignancy. A comprehensive understanding of the natural history, immune characteristics, and transplant outcomes has remained elusive. Here, in a multi-institutional global collaboration, we collected the clinical information of 49 patients from 29 families (CD27, n = 33; CD70, n = 16), including 24 previously unreported individuals and identified a total of 16 distinct mutations in CD27, and 8 in CD70, respectively. The majority of patients (90%) were EBV+ at diagnosis, but only ∼30% presented with infectious mononucleosis. Lymphoproliferation and lymphoma were the main clinical manifestations (70% and 43%, respectively), and 9 of the CD27-deficient patients developed HLH. Twenty-one patients (43%) developed autoinflammatory features including uveitis, arthritis, and periodic fever. Detailed immunological characterization revealed aberrant generation of memory B and T cells, including a paucity of EBV-specific T cells, and impaired effector function of CD8+ T cells, thereby providing mechanistic insight into cellular defects underpinning the clinical features of disrupted CD27/CD70 signaling. Nineteen patients underwent allogeneic hematopoietic stem cell transplantation (HSCT) prior to adulthood predominantly because of lymphoma, with 95% survival without disease recurrence. Our data highlight the marked predisposition to lymphoma of both CD27- and CD70-deficient patients. The excellent outcome after HSCT supports the timely implementation of this treatment modality particularly in patients presenting with malignant transformation to lymphoma.
- Published
- 2020
- Full Text
- View/download PDF
34. A guide to cancer immunotherapy: from T cell basic science to clinical practice.
- Author
-
Waldman AD, Fritz JM, and Lenardo MJ
- Subjects
- Animals, Cancer Vaccines therapeutic use, Clinical Trials as Topic, Humans, Immune Checkpoint Inhibitors, Immunotherapy, Adoptive, Immunotherapy trends, Neoplasms therapy, T-Lymphocytes immunology
- Abstract
The T lymphocyte, especially its capacity for antigen-directed cytotoxicity, has become a central focus for engaging the immune system in the fight against cancer. Basic science discoveries elucidating the molecular and cellular biology of the T cell have led to new strategies in this fight, including checkpoint blockade, adoptive cellular therapy and cancer vaccinology. This area of immunological research has been highly active for the past 50 years and is now enjoying unprecedented bench-to-bedside clinical success. Here, we provide a comprehensive historical and biological perspective regarding the advent and clinical implementation of cancer immunotherapeutics, with an emphasis on the fundamental importance of T lymphocyte regulation. We highlight clinical trials that demonstrate therapeutic efficacy and toxicities associated with each class of drug. Finally, we summarize emerging therapies and emphasize the yet to be elucidated questions and future promise within the field of cancer immunotherapy.
- Published
- 2020
- Full Text
- View/download PDF
35. HEM1 deficiency disrupts mTORC2 and F-actin control in inherited immunodysregulatory disease.
- Author
-
Cook SA, Comrie WA, Poli MC, Similuk M, Oler AJ, Faruqi AJ, Kuhns DB, Yang S, Vargas-Hernández A, Carisey AF, Fournier B, Anderson DE, Price S, Smelkinson M, Abou Chahla W, Forbes LR, Mace EM, Cao TN, Coban-Akdemir ZH, Jhangiani SN, Muzny DM, Gibbs RA, Lupski JR, Orange JS, Cuvelier GDE, Al Hassani M, Al Kaabi N, Al Yafei Z, Jyonouchi S, Raje N, Caldwell JW, Huang Y, Burkhardt JK, Latour S, Chen B, ElGhazali G, Rao VK, Chinn IK, and Lenardo MJ
- Subjects
- ADP-Ribosylation Factor 1 metabolism, CD4-Positive T-Lymphocytes immunology, Cell Proliferation, Humans, Immunologic Deficiency Syndromes immunology, Lymphoproliferative Disorders immunology, Membrane Proteins genetics, Pedigree, Phosphorylation, Wiskott-Aldrich Syndrome Protein Family chemistry, Wiskott-Aldrich Syndrome Protein Family metabolism, Actins metabolism, Cytokines biosynthesis, Immunologic Deficiency Syndromes genetics, Lymphoproliferative Disorders genetics, Mechanistic Target of Rapamycin Complex 2 metabolism, Membrane Proteins physiology
- Abstract
Immunodeficiency often coincides with hyperactive immune disorders such as autoimmunity, lymphoproliferation, or atopy, but this coincidence is rarely understood on a molecular level. We describe five patients from four families with immunodeficiency coupled with atopy, lymphoproliferation, and cytokine overproduction harboring mutations in NCKAP1L , which encodes the hematopoietic-specific HEM1 protein. These mutations cause the loss of the HEM1 protein and the WAVE regulatory complex (WRC) or disrupt binding to the WRC regulator, Arf1, thereby impairing actin polymerization, synapse formation, and immune cell migration. Diminished cortical actin networks caused by WRC loss led to uncontrolled cytokine release and immune hyperresponsiveness. HEM1 loss also blocked mechanistic target of rapamycin complex 2 (mTORC2)-dependent AKT phosphorylation, T cell proliferation, and selected effector functions, leading to immunodeficiency. Thus, the evolutionarily conserved HEM1 protein simultaneously regulates filamentous actin (F-actin) and mTORC2 signaling to achieve equipoise in immune responses., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
36. Human Plasma-like Medium Improves T Lymphocyte Activation.
- Author
-
Leney-Greene MA, Boddapati AK, Su HC, Cantor JR, and Lenardo MJ
- Abstract
T lymphocytes are critical for effective immunity, and the ability to study their behavior in vitro can facilitate major insights into their development, function, and fate. However, the composition of human plasma differs from conventional media, and we hypothesized that such differences could impact immune cell physiology. Here, we showed that relative to the medium typically used to culture lymphocytes (RPMI), a physiologic medium (human plasma-like medium; HPLM) induced markedly different transcriptional responses in human primary T cells and in addition, improved their activation upon antigen stimulation. We found that this medium-dependent effect on T cell activation is linked to Ca
2+ , which is six-fold higher in HPLM than in RPMI. Thus, a medium that more closely resembles human plasma has striking effects on T cell biology, further demonstrates that medium composition can profoundly affect experimental results, and broadly suggests that physiologic media may offer a valuable way to study cultured immune cells., Competing Interests: Declaration of Interests The authors declare no competing interests., (Published by Elsevier Inc.)- Published
- 2020
- Full Text
- View/download PDF
37. Defective glycosylation and multisystem abnormalities characterize the primary immunodeficiency XMEN disease.
- Author
-
Ravell JC, Matsuda-Lennikov M, Chauvin SD, Zou J, Biancalana M, Deeb SJ, Price S, Su HC, Notarangelo G, Jiang P, Morawski A, Kanellopoulou C, Binder K, Mukherjee R, Anibal JT, Sellers B, Zheng L, He T, George AB, Pittaluga S, Powers A, Kleiner DE, Kapuria D, Ghany M, Hunsberger S, Cohen JI, Uzel G, Bergerson J, Wolfe L, Toro C, Gahl W, Folio LR, Matthews H, Angelus P, Chinn IK, Orange JS, Trujillo-Vargas CM, Franco JL, Orrego-Arango J, Gutiérrez-Hincapié S, Patel NC, Raymond K, Patiroglu T, Unal E, Karakukcu M, Day AG, Mehta P, Masutani E, De Ravin SS, Malech HL, Altan-Bonnet G, Rao VK, Mann M, and Lenardo MJ
- Subjects
- Antigens, CD genetics, Antigens, CD immunology, Autoimmune Lymphoproliferative Syndrome genetics, Autoimmune Lymphoproliferative Syndrome pathology, CD4-CD8 Ratio, Cation Transport Proteins genetics, Cation Transport Proteins immunology, Female, Glycosylation, Humans, Magnesium Deficiency genetics, Magnesium Deficiency pathology, Male, X-Linked Combined Immunodeficiency Diseases genetics, X-Linked Combined Immunodeficiency Diseases pathology, Autoimmune Lymphoproliferative Syndrome immunology, Magnesium Deficiency immunology, X-Linked Combined Immunodeficiency Diseases immunology
- Abstract
X-linked immunodeficiency with magnesium defect, EBV infection, and neoplasia (XMEN) disease are caused by deficiency of the magnesium transporter 1 (MAGT1) gene. We studied 23 patients with XMEN, 8 of whom were EBV naive. We observed lymphadenopathy (LAD), cytopenias, liver disease, cavum septum pellucidum (CSP), and increased CD4-CD8-B220-TCRαβ+ T cells (αβDNTs), in addition to the previously described features of an inverted CD4/CD8 ratio, CD4+ T lymphocytopenia, increased B cells, dysgammaglobulinemia, and decreased expression of the natural killer group 2, member D (NKG2D) receptor. EBV-associated B cell malignancies occurred frequently in EBV-infected patients. We studied patients with XMEN and patients with autoimmune lymphoproliferative syndrome (ALPS) by deep immunophenotyping (32 immune markers) using time-of-flight mass cytometry (CyTOF). Our analysis revealed that the abundance of 2 populations of naive B cells (CD20+CD27-CD22+IgM+HLA-DR+CXCR5+CXCR4++CD10+CD38+ and CD20+CD27-CD22+IgM+HLA-DR+CXCR5+CXCR4+CD10-CD38-) could differentially classify XMEN, ALPS, and healthy individuals. We also performed glycoproteomics analysis on T lymphocytes and show that XMEN disease is a congenital disorder of glycosylation that affects a restricted subset of glycoproteins. Transfection of MAGT1 mRNA enabled us to rescue proteins with defective glycosylation. Together, these data provide new clinical and pathophysiological foundations with important ramifications for the diagnosis and treatment of XMEN disease.
- Published
- 2020
- Full Text
- View/download PDF
38. Mutations that prevent caspase cleavage of RIPK1 cause autoinflammatory disease.
- Author
-
Lalaoui N, Boyden SE, Oda H, Wood GM, Stone DL, Chau D, Liu L, Stoffels M, Kratina T, Lawlor KE, Zaal KJM, Hoffmann PM, Etemadi N, Shield-Artin K, Biben C, Tsai WL, Blake MD, Kuehn HS, Yang D, Anderton H, Silke N, Wachsmuth L, Zheng L, Moura NS, Beck DB, Gutierrez-Cruz G, Ombrello AK, Pinto-Patarroyo GP, Kueh AJ, Herold MJ, Hall C, Wang H, Chae JJ, Dmitrieva NI, McKenzie M, Light A, Barham BK, Jones A, Romeo TM, Zhou Q, Aksentijevich I, Mullikin JC, Gross AJ, Shum AK, Hawkins ED, Masters SL, Lenardo MJ, Boehm M, Rosenzweig SD, Pasparakis M, Voss AK, Gadina M, Kastner DL, and Silke J
- Subjects
- Animals, Caspase 3 metabolism, Female, Hereditary Autoinflammatory Diseases genetics, Hereditary Autoinflammatory Diseases pathology, Humans, MAP Kinase Kinase Kinases genetics, MAP Kinase Kinase Kinases metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Pedigree, Receptor-Interacting Protein Serine-Threonine Kinases deficiency, Receptor-Interacting Protein Serine-Threonine Kinases genetics, Caspase 8 metabolism, Hereditary Autoinflammatory Diseases metabolism, Mutation, Receptor-Interacting Protein Serine-Threonine Kinases metabolism
- Abstract
RIPK1 is a key regulator of innate immune signalling pathways. To ensure an optimal inflammatory response, RIPK1 is regulated post-translationally by well-characterized ubiquitylation and phosphorylation events, as well as by caspase-8-mediated cleavage
1-7 . The physiological relevance of this cleavage event remains unclear, although it is thought to inhibit activation of RIPK3 and necroptosis8 . Here we show that the heterozygous missense mutations D324N, D324H and D324Y prevent caspase cleavage of RIPK1 in humans and result in an early-onset periodic fever syndrome and severe intermittent lymphadenopathy-a condition we term 'cleavage-resistant RIPK1-induced autoinflammatory syndrome'. To define the mechanism for this disease, we generated a cleavage-resistant Ripk1D325A mutant mouse strain. Whereas Ripk1-/- mice died postnatally from systemic inflammation, Ripk1D325A/D325A mice died during embryogenesis. Embryonic lethality was completely prevented by the combined loss of Casp8 and Ripk3, but not by loss of Ripk3 or Mlkl alone. Loss of RIPK1 kinase activity also prevented Ripk1D325A/D325A embryonic lethality, although the mice died before weaning from multi-organ inflammation in a RIPK3-dependent manner. Consistently, Ripk1D325A/D325A and Ripk1D325A/+ cells were hypersensitive to RIPK3-dependent TNF-induced apoptosis and necroptosis. Heterozygous Ripk1D325A/+ mice were viable and grossly normal, but were hyper-responsive to inflammatory stimuli in vivo. Our results demonstrate the importance of caspase-mediated RIPK1 cleavage during embryonic development and show that caspase cleavage of RIPK1 not only inhibits necroptosis but also maintains inflammatory homeostasis throughout life.- Published
- 2020
- Full Text
- View/download PDF
39. Magnesium transporter 1 (MAGT1) deficiency causes selective defects in N- linked glycosylation and expression of immune-response genes.
- Author
-
Matsuda-Lennikov M, Biancalana M, Zou J, Ravell JC, Zheng L, Kanellopoulou C, Jiang P, Notarangelo G, Jing H, Masutani E, Oler AJ, Olano LR, Schulz BL, and Lenardo MJ
- Subjects
- Cation Transport Proteins genetics, Epstein-Barr Virus Infections genetics, Glycoproteins metabolism, Glycosylation, HEK293 Cells, Homeostasis, Humans, Killer Cells, Natural metabolism, Magnesium metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Mutation, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Cation Transport Proteins metabolism, Magnesium Deficiency genetics, Neoplasms genetics
- Abstract
Magnesium transporter 1 (MAGT1) critically mediates magnesium homeostasis in eukaryotes and is highly-conserved across different evolutionary branches. In humans, loss-of-function mutations in the MAGT1 gene cause X-linked magnesium deficiency with Epstein-Barr virus (EBV) infection and neoplasia (XMEN), a disease that has a broad range of clinical and immunological consequences. We have previously shown that EBV susceptibility in XMEN is associated with defective expression of the antiviral natural-killer group 2 member D (NKG2D) protein and abnormal Mg
2+ transport. New evidence suggests that MAGT1 is the human homolog of the yeast OST3/OST6 proteins that form an integral part of the N -linked glycosylation complex, although the exact contributions of these perturbations in the glycosylation pathway to disease pathogenesis are still unknown. Using MS-based glycoproteomics, along with CRISPR/Cas9-KO cell lines, natural killer cell-killing assays, and RNA-Seq experiments, we now demonstrate that humans lacking functional MAGT1 have a selective deficiency in both immune and nonimmune glycoproteins, and we identified several critical glycosylation defects in important immune-response proteins and in the expression of genes involved in immunity, particularly CD28. We show that MAGT1 function is partly interchangeable with that of the paralog protein tumor-suppressor candidate 3 (TUSC3) but that each protein has a different tissue distribution in humans. We observed that MAGT1-dependent glycosylation is sensitive to Mg2+ levels and that reduced Mg2+ impairs immune-cell function via the loss of specific glycoproteins. Our findings reveal that defects in protein glycosylation and gene expression underlie immune defects in an inherited disease due to MAGT1 deficiency., (© 2019 Matsuda-Lennikov et al.)- Published
- 2019
- Full Text
- View/download PDF
40. Mg 2+ regulation of kinase signaling and immune function.
- Author
-
Kanellopoulou C, George AB, Masutani E, Cannons JL, Ravell JC, Yamamoto TN, Smelkinson MG, Jiang PD, Matsuda-Lennikov M, Reilley J, Handon R, Lee PH, Miller JR, Restifo NP, Zheng L, Schwartzberg PL, Young M, and Lenardo MJ
- Subjects
- Animals, Biocatalysis drug effects, Blood Donors, CD4-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes drug effects, Calcium metabolism, Catalytic Domain drug effects, Cells, Cultured, Humans, Lymphocyte Activation drug effects, Magnesium blood, Magnesium chemistry, Male, Mice, Mice, Inbred C57BL, Orthomyxoviridae Infections blood, Orthomyxoviridae Infections virology, Osmolar Concentration, Protein Serine-Threonine Kinases metabolism, Protein-Tyrosine Kinases chemistry, Receptors, Antigen, T-Cell metabolism, Signal Transduction drug effects, Signal Transduction immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Influenza A Virus, H1N1 Subtype immunology, Magnesium pharmacology, Orthomyxoviridae Infections immunology, Protein-Tyrosine Kinases metabolism
- Abstract
Mg
2+ is required at micromolar concentrations as a cofactor for ATP, enzymatic reactions, and other biological processes. We show that decreased extracellular Mg2+ reduced intracellular Mg2+ levels and impaired the Ca2+ flux, activation marker up-regulation, and proliferation after T cell receptor (TCR) stimulation. Reduced Mg2+ specifically impairs TCR signal transduction by IL-2-inducible T cell kinase (ITK) due to a requirement for a regulatory Mg2+ in the catalytic pocket of ITK. We also show that altered catalytic efficiency by millimolar changes in free basal Mg2+ is an unrecognized but conserved feature of other serine/threonine and tyrosine kinases, suggesting a Mg2+ regulatory paradigm of kinase function. Finally, a reduced serum Mg2+ concentration in mice causes an impaired CD8+ T cell response to influenza A virus infection, reduces T cell activation, and exacerbates morbidity. Thus, Mg2+ directly regulates the active site of specific kinases during T cell responses, and maintaining a high serum Mg2+ concentration is important for antiviral immunity in otherwise healthy animals., (This is a work of the U.S. Government and is not subject to copyright protection in the United States. Foreign copyrights may apply.)- Published
- 2019
- Full Text
- View/download PDF
41. Development of immune checkpoint therapy for cancer.
- Author
-
Fritz JM and Lenardo MJ
- Subjects
- Animals, Combined Modality Therapy, Humans, Immune Tolerance, T-Lymphocytes immunology, Immunotherapy, Neoplasms immunology, Neoplasms therapy
- Abstract
Since the early 20th century, immunologists have investigated mechanisms that protect vertebrates from damaging immune responses against self-antigens by mature lymphocytes, i.e., peripheral tolerance. These mechanisms have been increasingly delineated at the molecular level, ultimately culminating in new therapeutics that have revolutionized clinical oncology. Here, we describe basic science and clinical discoveries that converge mainly on two molecules, CTLA-4 and PD-1, that were recognized with the 2018 Nobel Prize in Physiology or Medicine awarded to James Allison and Tasuku Honjo. We discuss their investigations and those of many others in the field that contravene tolerance through checkpoint inhibition to boost immune killing of malignant cells. We also discuss the mechanisms underlying each therapy, the efficacy achieved, and the complications of therapy. Finally, we hint at research questions for the future that could widen the success of cancer immunotherapy., (This is a work of the U.S. Government and is not subject to copyright protection in the United States. Foreign copyrights may apply.)
- Published
- 2019
- Full Text
- View/download PDF
42. Human interleukin-2 receptor β mutations associated with defects in immunity and peripheral tolerance.
- Author
-
Zhang Z, Gothe F, Pennamen P, James JR, McDonald D, Mata CP, Modis Y, Alazami AM, Acres M, Haller W, Bowen C, Döffinger R, Sinclair J, Brothers S, Zhang Y, Matthews HF, Naudion S, Pelluard F, Alajlan H, Yamazaki Y, Notarangelo LD, Thaventhiran JE, Engelhardt KR, Al-Mousa H, Hambleton S, Rooryck C, Smith KGC, and Lenardo MJ
- Subjects
- Alleles, Autoimmunity genetics, Genotype, HEK293 Cells, Humans, Immunologic Deficiency Syndromes genetics, Killer Cells, Natural metabolism, Lentivirus metabolism, Mutation, Missense genetics, Phenotype, Phosphorylation, STAT3 Transcription Factor metabolism, STAT5 Transcription Factor metabolism, Signal Transduction, T-Lymphocytes metabolism, Immune Tolerance genetics, Immunity genetics, Interleukin-2 Receptor beta Subunit genetics, Mutation genetics
- Abstract
Interleukin-2, which conveys essential signals for immunity, operates through a heterotrimeric receptor. Here we identify human interleukin-2 receptor (IL-2R) β chain ( IL2RB ) gene defects as a cause of life-threatening immune dysregulation. We report three homozygous mutations in the IL2RB gene of eight individuals from four consanguineous families that cause disease by distinct mechanisms. Nearly all patients presented with autoantibodies, hypergammaglobulinemia, bowel inflammation, dermatological abnormalities, lymphadenopathy, and cytomegalovirus disease. Patient T lymphocytes lacked surface expression of IL-2Rβ and were unable to respond to IL-2 stimulation. By contrast, natural killer cells retained partial IL-2Rβ expression and function. IL-2Rβ loss of function was recapitulated in a recombinant system in which IL2RB mutations caused reduced surface expression and IL-2 binding. Stem cell transplant ameliorated clinical symptoms in one patient; forced expression of wild-type IL-2Rβ also increased the IL-2 responsiveness of patient T lymphocytes in vitro. Insights from these patients can inform the development of IL-2-based therapeutics for immunological diseases and cancer., (This is a work of the U.S. Government and is not subject to copyright protection in the United States. Foreign copyrights may apply.)
- Published
- 2019
- Full Text
- View/download PDF
43. Introduction: Continuing insights into the healthy and diseased immune system through human genetic investigation.
- Author
-
Lenardo MJ and Holland SM
- Subjects
- Animals, Gene-Environment Interaction, Humans, Mutation genetics, Immune System, Immunologic Deficiency Syndromes genetics
- Published
- 2019
- Full Text
- View/download PDF
44. Erratum to "Plasma magnesium is inversely associated with Epstein-Barr virus load in peripheral blood and Burkitt lymphoma in Uganda" [Cancer Epidemiol. 52 (2018) 70-74].
- Author
-
Ravell J, Otim I, Nabalende H, Legason ID, Reynolds SJ, Ogwang MD, Ndugwa CM, Marshall V, Whitby D, Goedert JJ, Engels EA, Bhatia K, Lenardo MJ, and Mbulaiteye SM
- Published
- 2018
- Full Text
- View/download PDF
45. RELA haploinsufficiency in CD4 lymphoproliferative disease with autoimmune cytopenias.
- Author
-
Comrie WA, Faruqi AJ, Price S, Zhang Y, Rao VK, Su HC, and Lenardo MJ
- Subjects
- Child, Child, Preschool, Humans, Male, NF-kappa B immunology, Autoimmune Diseases immunology, Autoimmune Lymphoproliferative Syndrome immunology, CD4-Positive T-Lymphocytes immunology, Haploinsufficiency immunology, Lymphoproliferative Disorders immunology, Transcription Factor RelA immunology
- Published
- 2018
- Full Text
- View/download PDF
46. Plasma magnesium is inversely associated with Epstein-Barr virus load in peripheral blood and Burkitt lymphoma in Uganda.
- Author
-
Ravell J, Otim I, Nabalende H, Legason ID, Reynolds SJ, Ogwang MD, Ndugwa CM, Marshall V, Whitby D, Goedert JJ, Engels EA, Bhatia K, Lenardo MJ, and Mbulaiteye SM
- Subjects
- Adolescent, Adult, Burkitt Lymphoma diagnosis, Burkitt Lymphoma epidemiology, Child, Child, Preschool, Epstein-Barr Virus Infections virology, Female, Humans, Infant, Infant, Newborn, Male, Uganda epidemiology, Young Adult, Burkitt Lymphoma blood, Burkitt Lymphoma virology, Epstein-Barr Virus Infections complications, Herpesvirus 4, Human pathogenicity, Magnesium blood, Viral Load
- Abstract
Background: Epstein-Barr virus (EBV) causes endemic Burkitt lymphoma (eBL). EBV control was improved by magnesium (Mg
2+ ) supplementation in XMEN, an X-linked genetic disease associated with Mg2+ deficiency, high circulating EBV levels (viral loads), and EBV-related lymphomas. We, therefore, investigated the relationship between Mg2+ levels and EBV levels and eBL in Uganda., Methods: Plasma Mg2+ was measured in 45 women with low or high circulating EBV levels, 40 pediatric eBL cases, and 79 healthy children. Mg2+ uptake by T-lymphocytes was evaluated in samples from healthy donors., Results: Plasma Mg2+ deficiency (plasma level <1.8 mg/dl) was more likely in women with high- vs. low-EBV levels (76.0% vs. 35%; odds ratio [OR] 11.3, 95% CI 2.14-60.2), controlling for age, and in eBL cases than controls (42.0% vs. 13.9%; OR 3.61, 95% CI 1.32-9.88), controlling for sex, age group, and malaria status. Mg2+ uptake by T-lymphocytes was related to extracellular Mg2+ concentration., Interpretation: Plasma Mg2+ deficiency is associated with high EBV levels and eBL., (Published by Elsevier Ltd.)- Published
- 2018
- Full Text
- View/download PDF
47. Molecular Classification of Primary Immunodeficiencies of T Lymphocytes.
- Author
-
Comrie WA and Lenardo MJ
- Subjects
- Autoimmune Diseases immunology, Humans, Immune Tolerance, Signal Transduction, Autoimmunity, B-Lymphocytes immunology, Immunologic Deficiency Syndromes classification, Immunologic Deficiency Syndromes immunology, T-Lymphocytes immunology
- Abstract
Proper regulation of the immune system is required for protection against pathogens and preventing autoimmune disorders. Inborn errors of the immune system due to inherited or de novo germline mutations can lead to the loss of protective immunity, aberrant immune homeostasis, and the development of autoimmune disease, or combinations of these. Forward genetic screens involving clinical material from patients with primary immunodeficiencies (PIDs) can vary in severity from life-threatening disease affecting multiple cell types and organs to relatively mild disease with susceptibility to a limited range of pathogens or mild autoimmune conditions. As central mediators of innate and adaptive immune responses, T cells are critical orchestrators and effectors of the immune response. As such, several PIDs result from loss of or altered T cell function. PID-associated functional defects range from complete absence of T cell development to uncontrolled effector cell activation. Furthermore, the gene products of known PID causal genes are involved in diverse molecular pathways ranging from T cell receptor signaling to regulators of protein glycosylation. Identification of the molecular and biochemical cause of PIDs can not only guide the course of treatment for patients, but also inform our understanding of the basic biology behind T cell function. In this chapter, we review PIDs with known genetic causes that intrinsically affect T cell function with particular focus on perturbations of biochemical pathways., (2018 Published by Elsevier Inc.)
- Published
- 2018
- Full Text
- View/download PDF
48. STAT5B: A Differential Regulator of the Life and Death of CD4 + Effector Memory T Cells.
- Author
-
Majri SS, Fritz JM, Villarino AV, Zheng L, Kanellopoulou C, Chaigne-Delalande B, Grönholm J, Niemela JE, Afzali B, Biancalana M, Pittaluga S, Sun A, Cohen JL, Holland SM, O'Shea JJ, Uzel G, and Lenardo MJ
- Subjects
- Animals, Antibodies, Neutralizing metabolism, Autoimmune Lymphoproliferative Syndrome genetics, Cells, Cultured, Female, Humans, Immunologic Memory, Interleukin-2 immunology, Lymphocyte Activation, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutation, Missense genetics, STAT5 Transcription Factor genetics, Signal Transduction, Transcription, Genetic, Apoptosis, Autoimmune Lymphoproliferative Syndrome immunology, CD4-Positive T-Lymphocytes immunology, Cell Survival, STAT5 Transcription Factor metabolism
- Abstract
Understanding the control of Ag restimulation-induced T cell death (RICD), especially in cancer immunotherapy, where highly proliferating T cells will encounter potentially large amounts of tumor Ags, is important now more than ever. It has been known that growth cytokines make T cells susceptible to RICD, but the precise molecular mediators that govern this in T cell subsets is unknown until now. STAT proteins are a family of transcription factors that regulate gene expression programs underlying key immunological processes. In particular, STAT5 is known to favor the generation and survival of memory T cells. In this study, we report an unexpected role for STAT5 signaling in the death of effector memory T (TEM) cells in mice and humans. TEM cell death was prevented with neutralizing anti-IL-2 Ab or STAT5/JAK3 inhibitors, indicating that STAT5 signaling drives RICD in TEM cells. Moreover, we identified a unique patient with a heterozygous missense mutation in the coiled-coil domain of STAT5B that presented with autoimmune lymphoproliferative syndrome-like features. Similar to Stat5b
-/- mice, this patient exhibited increased CD4+ TEM cells in the peripheral blood. The mutant STAT5B protein dominantly interfered with STAT5-driven transcriptional activity, leading to global downregulation of STAT5-regulated genes in patient T cells upon IL-2 stimulation. Notably, CD4+ TEM cells from the patient were strikingly resistant to cell death by in vitro TCR restimulation, a finding that was recapitulated in Stat5b-/- mice. Hence, STAT5B is a crucial regulator of RICD in memory T cells in mice and humans.- Published
- 2018
- Full Text
- View/download PDF
49. Editorial overview: Autoimmunity: New genomics approaches are improving our understanding of autoimmunity.
- Author
-
Lo B and Lenardo MJ
- Subjects
- Humans, Inflammation genetics, Inflammation immunology, Autoimmune Diseases genetics, Autoimmune Diseases immunology, Autoimmunity genetics, Autoimmunity immunology
- Published
- 2017
- Full Text
- View/download PDF
50. Effective "activated PI3Kδ syndrome"-targeted therapy with the PI3Kδ inhibitor leniolisib.
- Author
-
Rao VK, Webster S, Dalm VASH, Šedivá A, van Hagen PM, Holland S, Rosenzweig SD, Christ AD, Sloth B, Cabanski M, Joshi AD, de Buck S, Doucet J, Guerini D, Kalis C, Pylvaenaeinen I, Soldermann N, Kashyap A, Uzel G, Lenardo MJ, Patel DD, Lucas CL, and Burkhart C
- Subjects
- Animals, Chemokines blood, Child, Child, Preschool, Class I Phosphatidylinositol 3-Kinases immunology, Class I Phosphatidylinositol 3-Kinases metabolism, Demography, Dose-Response Relationship, Drug, Female, Humans, Immunoglobulin M blood, Immunologic Deficiency Syndromes immunology, Immunologic Deficiency Syndromes pathology, Infant, Lymph Nodes drug effects, Lymph Nodes pathology, Lymphocyte Activation drug effects, Male, Mutation genetics, Organ Size, Phenotype, Primary Immunodeficiency Diseases, Pyridines pharmacokinetics, Pyrimidines pharmacokinetics, Rats, Spleen drug effects, Spleen pathology, T-Lymphocytes drug effects, T-Lymphocytes immunology, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases metabolism, Transfection, Class I Phosphatidylinositol 3-Kinases antagonists & inhibitors, Immunologic Deficiency Syndromes drug therapy, Immunologic Deficiency Syndromes enzymology, Molecular Targeted Therapy, Protein Kinase Inhibitors pharmacology, Pyridines pharmacology, Pyrimidines pharmacology
- Abstract
Pathogenic gain-of-function variants in the genes encoding phosphoinositide 3-kinase δ (PI3Kδ) lead to accumulation of transitional B cells and senescent T cells, lymphadenopathy, and immune deficiency (activated PI3Kδ syndrome [APDS]). Knowing the genetic etiology of APDS afforded us the opportunity to explore PI3Kδ inhibition as a precision-medicine therapy. Here, we report in vitro and in vivo effects of inhibiting PI3Kδ in APDS. Treatment with leniolisib (CDZ173), a selective PI3Kδ inhibitor, caused dose-dependent suppression of PI3Kδ pathway hyperactivation (measured as phosphorylation of AKT/S6) in cell lines ectopically expressing APDS-causative p110δ variants and in T-cell blasts derived from patients. A clinical trial with 6 APDS patients was conducted as a 12-week, open-label, multisite, within-subject, dose-escalation study of oral leniolisib to assess safety, pharmacokinetics, and effects on lymphoproliferation and immune dysregulation. Oral leniolisib led to a dose-dependent reduction in PI3K/AKT pathway activity assessed ex vivo and improved immune dysregulation. We observed normalization of circulating transitional and naive B cells, reduction in PD-1
+ CD4+ and senescent CD57+ CD4- T cells, and decreases in elevated serum immunoglobulin M and inflammatory markers including interferon γ, tumor necrosis factor, CXCL13, and CXCL10 with leniolisib therapy. After 12 weeks of treatment, all patients showed amelioration of lymphoproliferation with lymph node sizes and spleen volumes reduced by 39% (mean; range, 26%-57%) and 40% (mean; range, 13%-65%), respectively. Thus, leniolisib was well tolerated and improved laboratory and clinical parameters in APDS, supporting the specific inhibition of PI3Kδ as a promising new targeted therapy in APDS and other diseases characterized by overactivation of the PI3Kδ pathway. This trial was registered at www.clinicaltrials.gov as #NCT02435173.- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.