1
|
Zhang X, Ye Y, Sun W, Sheng Y, Kinoshita‐Ise M, Ito T, Lan C, Kwon O, Schaefer G, Wolk R, Hu S, Sun Q, Shen Y, Sakaki‐Yumoto M. Efficacy and safety of ritlecitinib in Asian patients with alopecia areata: A subgroup analysis of the ALLEGRO phase 2b/3 trial. J Dermatol 2025; 52:603-614. [PMID: 40071721 PMCID: PMC11975179 DOI: 10.1111/1346-8138.17539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/19/2024] [Indexed: 04/08/2025]
Abstract
This subgroup analysis of the ALLEGRO phase 2b/3 study (NCT3732807) assessed the efficacy and safety of multiple doses of ritlecitinib, an oral JAK3/TEC family kinase inhibitor, in Asian patients with alopecia areata (AA). Patients aged ≥12 years with AA and ≥50% scalp hair loss received once-daily ritlecitinib 50 or 30 mg (with or without 4-week 200-mg loading dose ["200/50" or "200/30"]) or 10 mg or placebo for 24 weeks, followed by a 24-week extension, in which patients initially assigned to placebo switched to 200/50 or 50 mg. In this subgroup analysis, Asian patients with response based on achieving a Severity of Alopecia Tool (SALT) score ≤20, SALT ≤10, ≥2-grade improvement or normal score on the eyebrow assessment (EBA) scale, and ≥2-grade improvement or normal score on the eyelash assessment (ELA) scale were evaluated through week 48. Safety was monitored throughout. In total, 186 Asian patients were randomized to ritlecitinib 200/50 mg (n = 33), 200/30 mg (n = 28), 50 mg (n = 43), 30 mg (n = 34), 10 mg (n = 17), placebo to 200/50 mg (n = 14), or placebo to 50 mg (n = 17). The proportions of patients treated with ritlecitinib ≥30 mg achieving a SALT score ≤20 response were 9.1%-36.4% at week 24 vs 0% for the 10-mg group and 3.2% for placebo. At week 48, 26.5%-55.6% of patients treated with ritlecitinib ≥30 mg achieved a SALT ≤20 response. At week 48, the proportions of patients treated with ritlecitinib ≥30 mg with EBA response were 41.9%-71.1% and with ELA response were 40.7%-57.9%. The most common adverse events were nasopharyngitis, folliculitis, upper respiratory tract infection, and urticaria. No serious or opportunistic infections, major adverse cardiovascular events, thromboembolic events, malignancies, or deaths were reported. Ritlecitinib demonstrated clinical efficacy and acceptable safety over 48 weeks in Asian patients ≥12 years with AA and ≥50% hair loss. Results for the Asian subpopulation were consistent with the overall population in the ALLEGRO-2b/3 study.
Collapse
Affiliation(s)
- Xingqi Zhang
- Department of DermatologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yanting Ye
- Department of DermatologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Weiling Sun
- The First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Youyu Sheng
- Huashan HospitalFudan UniversityShanghaiChina
| | | | - Taisuke Ito
- Hamamatsu University School of MedicineHamamatsuJapan
| | - Cheng‐Che Lan
- Department of DermatologyCollege of Medicine and Chung‐Ho Memorial Hospital, Kaohsiung Medical UniversityKaohsiungTaiwan
| | - Ohsang Kwon
- Seoul National University College of MedicineSeoulSouth Korea
| | | | | | | | | | | | | |
Collapse
|
2
|
Tziotzios C, Sinclair R, Lesiak A, Mehlis S, Kinoshita-Ise M, Tsianakas A, Luo X, Law EH, Ishowo-Adejumo R, Wolk R, Sadrarhami M, Lejeune A. Long-term safety and efficacy of ritlecitinib in adults and adolescents with alopecia areata and at least 25% scalp hair loss: Results from the ALLEGRO-LT phase 3, open-label study. J Eur Acad Dermatol Venereol 2025. [PMID: 39846397 DOI: 10.1111/jdv.20526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND ALLEGRO-LT is an ongoing, long-term, open-label, multicentre, phase 3 study of ritlecitinib in adults and adolescents with alopecia areata (AA). OBJECTIVES To evaluate ritlecitinib safety and efficacy through Month 24 in patients with AA and ≥25% scalp hair loss. METHODS ALLEGRO-LT enrolled rollover patients who previously received study intervention in either ALLEGRO phase 2a or 2b/3 studies and de novo patients who had not received treatment in either study. The de novo cohort results are reported here. Patients aged ≥12 years with AA and ≥25% scalp hair loss received a daily, 4-week 200-mg ritlecitinib loading dose, followed by daily 50-mg ritlecitinib. Analyses are based on data up to the cut-off (December 2022). Efficacy outcomes included proportions of patients achieving Severity of Alopecia Tool (SALT) scores ≤20 and ≤10, Patient Global Impression of Change (PGI-C) score of 'moderately improved' or 'greatly improved' and eyebrow assessment (EBA) and eyelash assessment (ELA) response (≥2-grade improvement from baseline or normal score in patients with abnormal baseline EBA/ELA). RESULTS Mean (SD) ritlecitinib exposure among the 449 de novo patients enrolled was 728.7 (273.81) days. At Month 24 (as observed), 73.5% and 66.4% of patients achieved SALT score ≤20 and ≤10; 82.4% had PGI-C response; 60.8% and 65.7% had EBA and ELA response. 86.1% of patients reported treatment-emergent adverse events (AEs); most were mild or moderate in severity, with the most frequent being positive SARS-CoV-2 test (24.2%), headache (20.8%) and pyrexia (13.0%). Rates of serious AEs, severe AEs and treatment discontinuations were 4.9%, 6.0% and 6.5%, respectively. Herpes zoster infection occurred in six patients, serious infections in four, malignancies (excluding nonmelanoma skin cancer) in three and major adverse cardiovascular events in three. CONCLUSIONS In patients with AA and ≥25% scalp hair loss, ritlecitinib demonstrated clinical efficacy and had an acceptable safety profile with long-term treatment. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov NCT04006457.
Collapse
Affiliation(s)
- C Tziotzios
- St John's Institute of Dermatology, King's College London, London, UK
| | - R Sinclair
- Sinclair Dermatology, Melbourne, Victoria, Australia
| | - A Lesiak
- Department of Dermatology, Pediatric Dermatology and Oncology, Medical University of Lodz, Lodz, Poland
- Laboratory of Autoinflammatory Genetic and Rare Skin Disorders, Department of Dermatology, Pediatric Dermatology and Oncology, Medical University of Lodz, Lodz, Poland
| | - S Mehlis
- Division of Dermatology, Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - M Kinoshita-Ise
- Department of Dermatology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - A Tsianakas
- Department of Dermatology, Fachklinik Bad Bentheim, Bad Bentheim, Germany
| | - X Luo
- Pfizer Inc, Groton, Connecticut, USA
| | - E H Law
- Pfizer Inc, New York City, New York, USA
| | | | - R Wolk
- Pfizer Inc, Groton, Connecticut, USA
| | | | | |
Collapse
|
3
|
Sinclair R, Law EH, Zhang X, Zhang F, Napatalung L, Zwillich SH, King B, Mesinkovska N. Patient-Reported Satisfaction with Hair Regrowth in a Study of Ritlecitinib in Alopecia Areata: Results from ALLEGRO-2b/3. Dermatology 2024; 240:767-777. [PMID: 38934147 PMCID: PMC11651339 DOI: 10.1159/000539536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Patients with alopecia areata (AA) report high levels of dissatisfaction with commonly used treatments. Patient-reported outcomes are essential to understanding patients' experiences with AA treatments. The objective of this study was to evaluate patient-reported satisfaction with hair growth among patients with AA receiving ritlecitinib or placebo and the correlation between clinician-assessed efficacy and patient-reported satisfaction. METHODS In the ALLEGRO-2b/3 (NCT03732807) trial, patients with AA and ≥50% scalp hair loss were randomized to daily ritlecitinib or placebo for 24 weeks, with a 24-week extension of continued ritlecitinib or switch from placebo to ritlecitinib. The Patient Satisfaction with Hair Growth (P-Sat) measure evaluated patients' satisfaction with hair growth in 3 domains: amount, quality, and overall satisfaction with hair growth. The prespecified analysis evaluated the proportion of patients who were slightly, moderately, or very satisfied with hair growth. Several post hoc analyses assessed the proportion of patients who were moderately/very satisfied and moderately/very dissatisfied and calculated polyserial correlations between change from baseline (CFB) in Severity of Alopecia Tool (SALT) and P-Sat scores at weeks 24 and 48. RESULTS At week 24, the proportion of patients (N = 718) reporting satisfaction (slightly, moderately, or very satisfied) overall with their hair growth ranged from 36.4% in the ritlecitinib 10-mg group (evaluated for dose ranging only) to 67.5% in the 200/50-mg group versus 22.6% in the placebo groups. In patients randomized to ritlecitinib, the proportion who were satisfied increased or was maintained at week 48. A substantially greater proportion of placebo patients who switched to ritlecitinib reported satisfaction at week 48 than at week 24. Similar results were observed for patient satisfaction with the amount and quality of hair growth. In the post hoc analyses defining satisfaction as moderately/very satisfied and dissatisfaction as moderately/very dissatisfied, the benefit of ritlecitinib was also observed. All P-Sat domain scores strongly correlated with CFB-SALT scores at weeks 24 (range 0.73-0.76; p < 0.05) and 48 (0.74-0.77; p < 0.05). CONCLUSIONS Patients receiving active ritlecitinib doses reported favorable results versus placebo in satisfaction with hair growth up to week 48. High concordance was observed between improvement in scalp hair growth evaluated by clinicians and patient-reported satisfaction.
Collapse
Affiliation(s)
| | | | - Xingqi Zhang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | - Lynne Napatalung
- Pfizer Inc., New York, NY, USA
- Department of Dermatology, Mount Sinai Hospital, New York, NY, USA
| | | | - Brett King
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
4
|
Stamenkovic V, Lautz JD, Harsh FM, Smith SEP. SRC family kinase inhibition rescues molecular and behavioral phenotypes, but not protein interaction network dynamics, in a mouse model of Fragile X syndrome. Mol Psychiatry 2024; 29:1392-1405. [PMID: 38297084 PMCID: PMC11524049 DOI: 10.1038/s41380-024-02418-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 02/02/2024]
Abstract
Glutamatergic synapses encode information from extracellular inputs using dynamic protein interaction networks (PINs) that undergo widespread reorganization following synaptic activity, allowing cells to distinguish between signaling inputs and generate coordinated cellular responses. Here, we investigate how Fragile X Messenger Ribonucleoprotein (FMRP) deficiency disrupts signal transduction through a glutamatergic synapse PIN downstream of NMDA receptor or metabotropic glutamate receptor (mGluR) stimulation. In cultured cortical neurons or acute cortical slices from P7, P17 and P60 FMR1-/y mice, the unstimulated protein interaction network state resembled that of wildtype littermates stimulated with mGluR agonists, demonstrating resting state pre-activation of mGluR signaling networks. In contrast, interactions downstream of NMDAR stimulation were similar to WT. We identified the Src family kinase (SFK) Fyn as a network hub, because many interactions involving Fyn were pre-activated in FMR1-/y animals. We tested whether targeting SFKs in FMR1-/y mice could modify disease phenotypes, and found that Saracatinib (SCB), an SFK inhibitor, normalized elevated basal protein synthesis, novel object recognition memory and social behavior in FMR1-/y mice. However, SCB treatment did not normalize the PIN to a wild-type-like state in vitro or in vivo, but rather induced extensive changes to protein complexes containing Shank3, NMDARs and Fyn. We conclude that targeting abnormal nodes of a PIN can identify potential disease-modifying drugs, but behavioral rescue does not correlate with PIN normalization.
Collapse
Affiliation(s)
- Vera Stamenkovic
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Felicia M Harsh
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
Ritmeester-Loy SA, Draper IH, Bueter EC, Lautz JD, Zhang-Wong Y, Gustafson JA, Wilson AL, Lin C, Gafken PR, Jensen MC, Orentas R, Smith SEP. Differential protein-protein interactions underlie signaling mediated by the TCR and a 4-1BB domain-containing CAR. Sci Signal 2024; 17:eadd4671. [PMID: 38442200 PMCID: PMC10986860 DOI: 10.1126/scisignal.add4671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/09/2024] [Indexed: 03/07/2024]
Abstract
Cells rely on activity-dependent protein-protein interactions to convey biological signals. For chimeric antigen receptor (CAR) T cells containing a 4-1BB costimulatory domain, receptor engagement is thought to stimulate the formation of protein complexes similar to those stimulated by T cell receptor (TCR)-mediated signaling, but the number and type of protein interaction-mediating binding domains differ between CARs and TCRs. Here, we performed coimmunoprecipitation mass spectrometry analysis of a second-generation, CD19-directed 4-1BB:ζ CAR (referred to as bbζCAR) and identified 128 proteins that increased their coassociation after target engagement. We compared activity-induced TCR and CAR signalosomes by quantitative multiplex coimmunoprecipitation and showed that bbζCAR engagement led to the activation of two modules of protein interactions, one similar to TCR signaling that was more weakly engaged by bbζCAR as compared with the TCR and one composed of TRAF signaling complexes that was not engaged by the TCR. Batch-to-batch and interindividual variations in production of the cytokine IL-2 correlated with differences in the magnitude of protein network activation. Future CAR T cell manufacturing protocols could measure, and eventually control, biological variation by monitoring these signalosome activation markers.
Collapse
Affiliation(s)
- Samuel A. Ritmeester-Loy
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Isabella H. Draper
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Eric C. Bueter
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Yue Zhang-Wong
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Joshua A. Gustafson
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, WA 98101 USA
| | - Ashley L. Wilson
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, WA 98101 USA
| | - Chenwei Lin
- Proteomics and Metabolomics Facility, Fred Hutchinson Cancer Center, Seattle, WA 98101, USA
| | - Philip R. Gafken
- Proteomics and Metabolomics Facility, Fred Hutchinson Cancer Center, Seattle, WA 98101, USA
| | - Michael C. Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, WA 98101 USA
- Department of Pediatrics, University of Washington, Seattle, WA 98101, USA
| | - Rimas Orentas
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98101, USA
| | - Stephen E. P. Smith
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98101, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98101, USA
| |
Collapse
|
6
|
Martin DA, Telliez JB, Pleasic-Williams S, Zhang Y, Tierney B, Blatnik M, Gale JD, Banfield C, Zhou Y, Lejeune A, Zwillich SH, Stevens E, Tiwari N, Kieras E, Karanam A. Target Occupancy and Functional Inhibition of JAK3 and TEC Family Kinases by Ritlecitinib in Healthy Adults: An Open-Label, Phase 1 Study. J Clin Pharmacol 2024; 64:67-79. [PMID: 37691236 DOI: 10.1002/jcph.2347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Abstract
Ritlecitinib is a small molecule in clinical development that covalently and irreversibly inhibits Janus kinase 3 (JAK3) and the TEC family of kinases (BTK, BMX, ITK, TXK, and TEC). This phase 1, open-label, parallel-group study assessed target occupancy and functional effects of ritlecitinib on JAK3 and TEC family kinases in healthy participants aged 18-60 years who received 50 or 200 mg single doses of ritlecitinib on day 1. Blood samples to assess ritlecitinib pharmacokinetics, target occupancy, and pharmacodynamics were collected over 48 hours. Target occupancy was assessed using mass spectroscopy. Functional inhibition of JAK3-dependent signaling was measured by the inhibition of the phosphorylation of its downstream target signal transducer and activator of transcription 5 (pSTAT5), following activation by interleukin 15 (IL-15). The functional inhibition of Bruton's tyrosine kinase (BTK)-dependent signaling was measured by the reduction in the upregulation of cluster of differentiation 69 (CD69), an early marker of B-cell activation, following treatment with anti-immunoglobulin D. Eight participants received one 50 mg ritlecitinib dose and 8 participants received one 200 mg dose. Ritlecitinib plasma exposure increased in an approximately dose-proportional manner from 50 to 200 mg. The maximal median JAK3 target occupancy was 72% for 50 mg and 64% for 200 mg. Ritlecitinib 50 mg had >94% maximal target occupancy of all TEC kinases, except BMX (87%), and 200 mg had >97% for all TEC kinases. For BTK and TEC, ritlecitinib maintained high target occupancy throughout a period of 48 hours. Ritlecitinib reduced pSTAT5 levels following IL-15- and BTK-dependent signaling in a dose-dependent manner. These target occupancy and functional assays demonstrate the dual inhibition of the JAK3- and BTK-dependent pathways by ritlecitinib. Further studies are needed to understand the contribution to clinical effects of inhibiting these pathways.
Collapse
|
7
|
Passeron T, King B, Seneschal J, Steinhoff M, Jabbari A, Ohyama M, Tobin DJ, Randhawa S, Winkler A, Telliez JB, Martin D, Lejeune A. Inhibition of T-cell activity in alopecia areata: recent developments and new directions. Front Immunol 2023; 14:1243556. [PMID: 38022501 PMCID: PMC10657858 DOI: 10.3389/fimmu.2023.1243556] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Alopecia areata (AA) is an autoimmune disease that has a complex underlying immunopathogenesis characterized by nonscarring hair loss ranging from small bald patches to complete loss of scalp, face, and/or body hair. Although the etiopathogenesis of AA has not yet been fully characterized, immune privilege collapse at the hair follicle (HF) followed by T-cell receptor recognition of exposed HF autoantigens by autoreactive cytotoxic CD8+ T cells is now understood to play a central role. Few treatment options are available, with the Janus kinase (JAK) 1/2 inhibitor baricitinib (2022) and the selective JAK3/tyrosine kinase expressed in hepatocellular carcinoma (TEC) inhibitor ritlecitinib (2023) being the only US Food and Drug Administration-approved systemic medications thus far for severe AA. Several other treatments are used off-label with limited efficacy and/or suboptimal safety and tolerability. With an increased understanding of the T-cell-mediated autoimmune and inflammatory pathogenesis of AA, additional therapeutic pathways beyond JAK inhibition are currently under investigation for the development of AA therapies. This narrative review presents a detailed overview about the role of T cells and T-cell-signaling pathways in the pathogenesis of AA, with a focus on those pathways targeted by drugs in clinical development for the treatment of AA. A detailed summary of new drugs targeting these pathways with expert commentary on future directions for AA drug development and the importance of targeting multiple T-cell-signaling pathways is also provided in this review.
Collapse
Affiliation(s)
- Thierry Passeron
- University Côte d’Azur, Centre Hospitalier Universitaire Nice, Department of Dermatology, Nice, France
- University Côte d’Azur, INSERM, U1065, C3M, Nice, France
| | - Brett King
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, United States
| | - Julien Seneschal
- Department of Dermatology and Paediatric Dermatology, National Reference Centre for Rare Skin Diseases, Saint-André Hospital, University of Bordeaux, Bordeaux, France
- Bordeaux University, Centre national de la recherche scientifique (CNRS), ImmunoConcept, UMR5164, Bordeaux, France
| | - Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology and Venereology, Weill Cornell Medicine-Qatar, Doha, Qatar
- College of Medicine, Qatar University, Doha, Qatar
- Department of Dermatology, Weill Cornell Medicine, New York, NY, United States
- College of Health and Life Sciences, Hamad Bin Khalifa University-Qatar, Doha, Qatar
| | - Ali Jabbari
- Department of Dermatology, University of Iowa, Iowa City, IA, United States
- Iowa City VA Medical Center, Iowa City, IA, United States
| | - Manabu Ohyama
- Department of Dermatology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Desmond J. Tobin
- Charles Institute of Dermatology, UCD School of Medicine, University College Dublin, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|
8
|
Wehle DT, Bass CS, Sulc J, Mirzaa G, Smith SEP. Protein interaction network analysis of mTOR signaling reveals modular organization. J Biol Chem 2023; 299:105271. [PMID: 37741456 PMCID: PMC10594569 DOI: 10.1016/j.jbc.2023.105271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is a serine-threonine kinase that acts as a central mediator of translation and plays important roles in cell growth, synaptic plasticity, cancer, and a wide range of developmental disorders. The signaling cascade linking lipid kinases (phosphoinositide 3-kinases), protein kinases (AKT), and translation initiation complexes (EIFs) to mTOR has been extensively modeled, but does not fully describe mTOR system behavior. Here, we use quantitative multiplex coimmunoprecipitation to monitor a protein interaction network (PIN) composed of 300+ binary interactions among mTOR-related proteins. Using a simple model system of serum-deprived or fresh-media-fed mouse 3T3 fibroblasts, we observed extensive PIN remodeling involving 27+ individual protein interactions after 1 h, despite phosphorylation changes observed after only 5 min. Using small molecule inhibitors of phosphoinositide 3-kinase, AKT, mTOR, MEK and ERK, we define subsets of the PIN, termed "modules", that respond differently to each inhibitor. Using primary fibroblasts from individuals with overgrowth disorders caused by pathogenic PIK3CA or MTOR variants, we find that hyperactivation of mTOR pathway components is reflected in a hyperactive PIN. Our data define a "modular" organization of the mTOR PIN in which coordinated groups of interactions respond to the activation or inhibition of distinct nodes, and demonstrate that kinase inhibitors affect the modular network architecture in a complex manner, inconsistent with simple linear models of signal transduction.
Collapse
Affiliation(s)
- Devin T Wehle
- Graduate Program in Neuroscience, University of Washington, Seattle, Washington, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Carter S Bass
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Josef Sulc
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Ghayda Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA; Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Stephen E P Smith
- Graduate Program in Neuroscience, University of Washington, Seattle, Washington, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA.
| |
Collapse
|
9
|
Wehle DT, Bass CS, Sulc J, Mirzaa G, Smith SEP. Protein interaction network analysis of mTOR signaling reveals modular organization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.552011. [PMID: 37577705 PMCID: PMC10418199 DOI: 10.1101/2023.08.04.552011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The mammalian target of rapamycin (mTOR) is a serine-threonine kinase that acts as a central mediator of translation, and plays important roles in cell growth, synaptic plasticity, cancer, and a wide range of developmental disorders. The signaling cascade linking lipid kinases (PI3Ks), protein kinases (AKT) and translation initiation complexes (EIFs) to mTOR has been extensively modeled, but does not fully describe mTOR system behavior. Here, we use quantitative multiplex co-immunoprecipitation to monitor a protein interaction network (PIN) composed of 300+ binary interactions among mTOR-related proteins. Using a simple model system of serum deprived or fresh-media-fed mouse 3T3 fibroblasts, we observed extensive PIN remodeling involving 27+ individual protein interactions after one hour, despite phosphorylation changes observed after only five minutes. Using small molecule inhibitors of PI3K, AKT, mTOR, MEK and ERK, we define subsets of the PIN, termed 'modules', that respond differently to each inhibitor. Using primary fibroblasts from individuals with overgrowth disorders caused by pathogenic PIK3CA or MTOR variants, we find that hyperactivation of mTOR pathway components is reflected in a hyperactive PIN. Our data define a "modular" organization of the mTOR PIN in which coordinated groups of interactions respond to activation or inhibition of distinct nodes, and demonstrate that kinase inhibitors affect the modular network architecture in a complex manner, inconsistent with simple linear models of signal transduction.
Collapse
Affiliation(s)
- Devin T Wehle
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Carter S Bass
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Josef Sulc
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ghayda Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Stephen E P Smith
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
10
|
King B, Zhang X, Harcha WG, Szepietowski JC, Shapiro J, Lynde C, Mesinkovska NA, Zwillich SH, Napatalung L, Wajsbrot D, Fayyad R, Freyman A, Mitra D, Purohit V, Sinclair R, Wolk R. Efficacy and safety of ritlecitinib in adults and adolescents with alopecia areata: a randomised, double-blind, multicentre, phase 2b-3 trial. Lancet 2023; 401:1518-1529. [PMID: 37062298 DOI: 10.1016/s0140-6736(23)00222-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/20/2022] [Accepted: 01/24/2023] [Indexed: 04/18/2023]
Abstract
BACKGROUND Alopecia areata is characterised by non-scarring loss of scalp, face, or body hair. We investigated the efficacy and safety of ritlecitinib, an oral, selective dual JAK3/TEC family kinase inhibitor, in patients with alopecia areata. METHODS In this randomised, double-blind, multicentre, phase 2b-3 trial done at 118 sites in 18 countries, patients aged 12 years and older with alopecia areata and at least 50% scalp hair loss were randomly assigned to oral ritlecitinib or placebo once-daily for 24 weeks, with or without a 4-week loading dose (50 mg, 30 mg, 10 mg, 200 mg loading dose followed by 50 mg, or 200 mg loading dose followed by 30 mg), followed by a 24-week extension period during which ritlecitinib groups continued their assigned doses and patients initially assigned to placebo switched to ritlecitinib 50 mg or 200 mg loading dose followed by 50 mg. Randomisation was done by use of an interactive response system and was stratified by baseline disease severity and age. The sponsor, patients, and investigators were masked to treatment, and all patients received the same number of tablets to maintain masking. The primary endpoint was Severity of Alopecia Tool (SALT) score 20 or less at week 24. The primary endpoint was assessed in all assigned patients, regardless of whether they received treatment. This study was registered with ClinicalTrials.gov, NCT03732807. FINDINGS Between Dec 3, 2018, and June 24, 2021, 1097 patients were screened and 718 were randomly assigned to receive ritlecitinib 200 mg + 50 mg (n=132), 200 mg + 30 mg (n=130), 50 mg (n=130), 30 mg (n=132), 10 mg (n=63), placebo to 50 mg (n=66), or placebo to 200 mg + 50 mg (n=65). 446 (62%) of 718 patients were female and 272 (38%) were male. 488 (68%) were White, 186 (26%) were Asian, and 27 (4%) were Black or African American. Of 718 patients randomly assigned, 104 patients discontinued treatment (34 withdrew, 19 adverse events [AEs], 12 physician decision, 12 lack of efficacy, 13 lost to follow up, five rolled over to long-term study transfer, four pregnancies, two protocol deviations, one declined to attend follow-up due to COVID-19, one attended last visit very late due to COVID-19, and one non-compliance). At week 24, 38 (31%) of 124 patients in the ritlecitinib 200 mg + 50 mg group, 27 (22%) of 121 patients in the 200 mg + 30 mg group, 29 (23%) of 124 patients in the 50 mg group, 17 (14%) of 119 patients in the 30 mg group, and two (2%) of 130 patients in the placebo group had a response based on SALT score 20 or less. The difference in response rate based on SALT score 20 or less between the placebo and the ritlecitinib 200 mg + 50 mg group was 29·1% (95% CI 21·2-37·9; p<0·0001), 20·8% (13·7-29·2; p<0·0001) for the 200 mg + 30 mg group, 21·9% (14·7-30·2; p<0·0001) for the 50 mg group, and 12·8% (6·7-20·4; p=0·0002) for the 30 mg group. Up to week 48 and including the follow-up period, AEs had been reported in 108 (82%) of 131 patients in the ritlecitinib 200 mg + 50 mg group, 105 (81%) of 129 patients in the 200 mg + 30 mg group, 110 (85%) of 130 patients in the 50 mg group, 106 (80%) of 132 patients in the 30 mg group, 47 (76%) of 62 patients in the 10 mg group, 54 (83%) of 65 patients placebo to ritlecitinib 200 mg + 50 mg in the extension period, and 57 (86%) of 66 patients in the placebo to 50 mg group. The incidence of each AE was similar between groups, and there were no deaths. INTERPRETATION Ritlecitinib was effective and well tolerated in patients aged 12 years and older with alopecia areata. Ritlecitinib might be a suitable treatment option for alopecia areata in patients who are candidates for systemic therapy. FUNDING Pfizer.
Collapse
Affiliation(s)
- Brett King
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA.
| | - Xingqi Zhang
- Department of Dermatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | - Jacek C Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Jerry Shapiro
- Department of Dermatology, New York University School of Medicine, New York, NY, USA
| | - Charles Lynde
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Natasha A Mesinkovska
- Department of Dermatology and Dermatopathology, School of Medicine, University of California, Irvine, CA, USA
| | | | - Lynne Napatalung
- Pfizer, New York, NY, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Teixeiro E, Daniels MA. In Vitro Analysis of Thymocyte Signaling. Methods Mol Biol 2023; 2580:303-313. [PMID: 36374466 DOI: 10.1007/978-1-0716-2740-2_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
When a developing thymocyte expresses a TCR, it is subjected to numerous interactions with self-peptide/MHC complexes that determine its fate. These include death by neglect, negative selection (apoptosis and lineage deviation), positive selection, and lineage commitment. Identifying signals that govern these unique cell fates requires the ability to assess the activity, level of expression, subcellular location, and molecular associations between numerous proteins within the developing T cell. Given the unique, temporal, and developmental changes that occur during development, isolating and analyzing small populations of thymocytes are necessary to get a complete picture of the development process. Thus, this chapter describes methods designed to analyze thymocyte signaling under various types of peptide-based stimulation in vitro.
Collapse
Affiliation(s)
- Emma Teixeiro
- Department of Molecular Microbiology and Immunology, NextGen Precision Health, University of Missouri, Columbia, MO, USA
| | - Mark A Daniels
- Department of Molecular Microbiology and Immunology, NextGen Precision Health, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
12
|
Lautz JD, Tsegay KB, Zhu Z, Gniffke EP, Welsh JP, Smith SEP. Synaptic protein interaction networks encode experience by assuming stimulus-specific and brain-region-specific states. Cell Rep 2021; 37:110076. [PMID: 34852231 PMCID: PMC8722361 DOI: 10.1016/j.celrep.2021.110076] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/01/2021] [Accepted: 11/09/2021] [Indexed: 11/02/2022] Open
Abstract
A core network of widely expressed proteins within the glutamatergic post-synapse mediates activity-dependent synaptic plasticity throughout the brain, but the specific proteomic composition of synapses differs between brain regions. Here, we address the question, how does proteomic composition affect activity-dependent protein-protein interaction networks (PINs) downstream of synaptic activity? Using quantitative multiplex co-immunoprecipitation, we compare the PIN response of in vivo or ex vivo neurons derived from different brain regions to activation by different agonists or different forms of eyeblink conditioning. We report that PINs discriminate between incoming stimuli using differential kinetics of overlapping and non-overlapping PIN parameters. Further, these "molecular logic rules" differ by brain region. We conclude that although the PIN of the glutamatergic post-synapse is expressed widely throughout the brain, its activity-dependent dynamics show remarkable stimulus-specific and brain-region-specific diversity. This diversity may help explain the challenges in developing molecule-specific drug therapies for neurological disorders.
Collapse
Affiliation(s)
- Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kaleb B Tsegay
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Zhiyi Zhu
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Edward P Gniffke
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - John P Welsh
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA.
| |
Collapse
|
13
|
Negraes PD, Trujillo CA, Yu NK, Wu W, Yao H, Liang N, Lautz JD, Kwok E, McClatchy D, Diedrich J, de Bartolome SM, Truong J, Szeto R, Tran T, Herai RH, Smith SEP, Haddad GG, Yates JR, Muotri AR. Altered network and rescue of human neurons derived from individuals with early-onset genetic epilepsy. Mol Psychiatry 2021; 26:7047-7068. [PMID: 33888873 PMCID: PMC8531162 DOI: 10.1038/s41380-021-01104-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 02/02/2023]
Abstract
Early-onset epileptic encephalopathies are severe disorders often associated with specific genetic mutations. In this context, the CDKL5 deficiency disorder (CDD) is a neurodevelopmental condition characterized by early-onset seizures, intellectual delay, and motor dysfunction. Although crucial for proper brain development, the precise targets of CDKL5 and its relation to patients' symptoms are still unknown. Here, induced pluripotent stem cells derived from individuals deficient in CDKL5 protein were used to generate neural cells. Proteomic and phosphoproteomic approaches revealed disruption of several pathways, including microtubule-based processes and cytoskeleton organization. While CDD-derived neural progenitor cells have proliferation defects, neurons showed morphological alterations and compromised glutamatergic synaptogenesis. Moreover, the electrical activity of CDD cortical neurons revealed hyperexcitability during development, leading to an overly synchronized network. Many parameters of this hyperactive network were rescued by lead compounds selected from a human high-throughput drug screening platform. Our results enlighten cellular, molecular, and neural network mechanisms of genetic epilepsy that could ultimately promote novel therapeutic opportunities for patients.
Collapse
Affiliation(s)
- Priscilla D Negraes
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Cleber A Trujillo
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
| | - Nam-Kyung Yu
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Wei Wu
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Hang Yao
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Nicholas Liang
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| | - Ellius Kwok
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Daniel McClatchy
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Jolene Diedrich
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Justin Truong
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Ryan Szeto
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Timothy Tran
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Roberto H Herai
- Experimental Multiuser Laboratory, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| | - Gabriel G Haddad
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Alysson R Muotri
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA, USA.
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA, USA.
| |
Collapse
|
14
|
Heavner WE, Lautz JD, Speed HE, Gniffke EP, Immendorf KB, Welsh JP, Baertsch NA, Smith SEP. Remodeling of the Homer-Shank interactome mediates homeostatic plasticity. Sci Signal 2021; 14:14/681/eabd7325. [PMID: 33947797 DOI: 10.1126/scisignal.abd7325] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neurons maintain stable levels of excitability using homeostatic synaptic scaling, which adjusts the strength of a neuron's postsynaptic inputs to compensate for extended changes in overall activity. Here, we investigated whether prolonged changes in activity affect network-level protein interactions at the synapse. We assessed a glutamatergic synapse protein interaction network (PIN) composed of 380 binary associations among 21 protein members in mouse neurons. Manipulating the activation of cultured mouse cortical neurons induced widespread bidirectional PIN alterations that reflected rapid rearrangements of glutamate receptor associations involving synaptic scaffold remodeling. Sensory deprivation of the barrel cortex in live mice (by whisker trimming) caused specific PIN rearrangements, including changes in the association between the glutamate receptor mGluR5 and the kinase Fyn. These observations are consistent with emerging models of experience-dependent plasticity involving multiple types of homeostatic responses. However, mice lacking Homer1 or Shank3B did not undergo normal PIN rearrangements, suggesting that the proteins encoded by these autism spectrum disorder-linked genes serve as structural hubs for synaptic homeostasis. Our approach demonstrates how changes in the protein content of synapses during homeostatic plasticity translate into functional PIN alterations that mediate changes in neuron excitability.
Collapse
Affiliation(s)
- Whitney E Heavner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Haley E Speed
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Edward P Gniffke
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Karen B Immendorf
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - John P Welsh
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA.,University of Washington Autism Center, Seattle, WA 98195, USA.,Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
| | - Nathan A Baertsch
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA. .,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA.,Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
15
|
Trujillo CA, Rice ES, Schaefer NK, Chaim IA, Wheeler EC, Madrigal AA, Buchanan J, Preissl S, Wang A, Negraes PD, Szeto RA, Herai RH, Huseynov A, Ferraz MSA, Borges FS, Kihara AH, Byrne A, Marin M, Vollmers C, Brooks AN, Lautz JD, Semendeferi K, Shapiro B, Yeo GW, Smith SEP, Green RE, Muotri AR. Reintroduction of the archaic variant of NOVA1 in cortical organoids alters neurodevelopment. Science 2021; 371:371/6530/eaax2537. [PMID: 33574182 DOI: 10.1126/science.aax2537] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/27/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022]
Abstract
The evolutionarily conserved splicing regulator neuro-oncological ventral antigen 1 (NOVA1) plays a key role in neural development and function. NOVA1 also includes a protein-coding difference between the modern human genome and Neanderthal and Denisovan genomes. To investigate the functional importance of an amino acid change in humans, we reintroduced the archaic allele into human induced pluripotent cells using genome editing and then followed their neural development through cortical organoids. This modification promoted slower development and higher surface complexity in cortical organoids with the archaic version of NOVA1 Moreover, levels of synaptic markers and synaptic protein coassociations correlated with altered electrophysiological properties in organoids expressing the archaic variant. Our results suggest that the human-specific substitution in NOVA1, which is exclusive to modern humans since divergence from Neanderthals, may have had functional consequences for our species' evolution.
Collapse
Affiliation(s)
- Cleber A Trujillo
- Department of Pediatrics and Department of Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA 92037, USA
| | - Edward S Rice
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Nathan K Schaefer
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Isaac A Chaim
- Department of Cellular & Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Emily C Wheeler
- Department of Cellular & Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Assael A Madrigal
- Department of Cellular & Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Justin Buchanan
- Department of Cellular & Molecular Medicine, Center for Epigenomics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sebastian Preissl
- Department of Cellular & Molecular Medicine, Center for Epigenomics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Allen Wang
- Department of Cellular & Molecular Medicine, Center for Epigenomics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Priscilla D Negraes
- Department of Pediatrics and Department of Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA 92037, USA
| | - Ryan A Szeto
- Department of Pediatrics and Department of Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA 92037, USA
| | - Roberto H Herai
- Experimental Multiuser Laboratory (LEM), Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil
| | - Alik Huseynov
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Mariana S A Ferraz
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-070, Brazil
| | - Fernando S Borges
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-070, Brazil
| | - Alexandre H Kihara
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-070, Brazil
| | - Ashley Byrne
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Maximillian Marin
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Christopher Vollmers
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Angela N Brooks
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Department of Pediatrics and Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
| | - Katerina Semendeferi
- Department of Anthropology, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA 92037, USA
| | - Beth Shapiro
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Howard Hughes Medical Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Gene W Yeo
- Department of Cellular & Molecular Medicine, Center for Epigenomics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Department of Pediatrics and Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
| | - Richard E Green
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Alysson R Muotri
- Department of Pediatrics and Department of Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
16
|
Neier SC, Ferrer A, Wilton KM, Smith SEP, Kelcher AMH, Pavelko KD, Canfield JM, Davis TR, Stiles RJ, Chen Z, McCluskey J, Burrows SR, Rossjohn J, Hebrink DM, Carmona EM, Limper AH, Kappes DJ, Wettstein PJ, Johnson AJ, Pease LR, Daniels MA, Neuhauser C, Gil D, Schrum AG. The early proximal αβ TCR signalosome specifies thymic selection outcome through a quantitative protein interaction network. Sci Immunol 2020; 4:4/32/eaal2201. [PMID: 30770409 DOI: 10.1126/sciimmunol.aal2201] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/17/2019] [Indexed: 12/18/2022]
Abstract
During αβ T cell development, T cell antigen receptor (TCR) engagement transduces biochemical signals through a protein-protein interaction (PPI) network that dictates dichotomous cell fate decisions. It remains unclear how signal specificity is communicated, instructing either positive selection to advance cell differentiation or death by negative selection. Early signal discrimination might occur by PPI signatures differing qualitatively (customized, unique PPI combinations for each signal), quantitatively (graded amounts of a single PPI series), or kinetically (speed of PPI pathway progression). Using a novel PPI network analysis, we found that early TCR-proximal signals distinguishing positive from negative selection appeared to be primarily quantitative in nature. Furthermore, the signal intensity of this PPI network was used to find an antigen dose that caused a classic negative selection ligand to induce positive selection of conventional αβ T cells, suggesting that the quantity of TCR triggering was sufficient to program selection outcome. Because previous work had suggested that positive selection might involve a qualitatively unique signal through CD3δ, we reexamined the block in positive selection observed in CD3δ0 mice. We found that CD3δ0 thymocytes were inhibited but capable of signaling positive selection, generating low numbers of MHC-dependent αβ T cells that expressed diverse TCR repertoires and participated in immune responses against infection. We conclude that the major role for CD3δ in positive selection is to quantitatively boost the signal for maximal generation of αβ T cells. Together, these data indicate that a quantitative network signaling mechanism through the early proximal TCR signalosome determines thymic selection outcome.
Collapse
Affiliation(s)
- Steven C Neier
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Alejandro Ferrer
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Katelynn M Wilton
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA.,Medical Scientist Training Program, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Stephen E P Smith
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - April M H Kelcher
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jenna M Canfield
- Molecular Pathogenesis and Therapeutics PhD Graduate Program, University of Missouri, Columbia, MO, USA
| | - Tessa R Davis
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Robert J Stiles
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Zhenjun Chen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Scott R Burrows
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia.,School of Medicine, University of Queensland, Brisbane, Queensland 4006, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Deanne M Hebrink
- Thoracic Diseases Research Unit, Division of Pulmonary Critical Care and Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Eva M Carmona
- Thoracic Diseases Research Unit, Division of Pulmonary Critical Care and Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Andrew H Limper
- Thoracic Diseases Research Unit, Division of Pulmonary Critical Care and Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Dietmar J Kappes
- Blood Cell Development and Cancer Keystone, Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Peter J Wettstein
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Larry R Pease
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mark A Daniels
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.,Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, USA
| | | | - Diana Gil
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA. .,Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, USA.,Department of Bioengineering, College of Engineering, University of Missouri, Columbia, MO, USA
| | - Adam G Schrum
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA. .,Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, USA.,Department of Bioengineering, College of Engineering, University of Missouri, Columbia, MO, USA
| |
Collapse
|
17
|
Brown EA, Neier SC, Neuhauser C, Schrum AG, Smith SEP. Quantification of Protein Interaction Network Dynamics using Multiplexed Co-Immunoprecipitation. J Vis Exp 2019. [PMID: 31498315 DOI: 10.3791/60029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dynamic protein-protein interactions control cellular behavior, from motility to DNA replication to signal transduction. However, monitoring dynamic interactions among multiple proteins in a protein interaction network is technically difficult. Here, we present a protocol for Quantitative Multiplex Immunoprecipitation (QMI), which allows quantitative assessment of fold changes in protein interactions based on relative fluorescence measurements of Proteins in Shared Complexes detected by Exposed Surface epitopes (PiSCES). In QMI, protein complexes from cell lysates are immunoprecipitated onto microspheres, and then probed with a labeled antibody for a different protein in order to quantify the abundance of PiSCES. Immunoprecipitation antibodies are conjugated to different MagBead spectral regions, which allows a flow cytometer to differentiate multiple parallel immunoprecipitations and simultaneously quantify the amount of probe antibody associated with each. QMI does not require genetic tagging and can be performed using minimal biomaterial compared to other immunoprecipitation methods. QMI can be adapted for any defined group of interacting proteins, and has thus far been used to characterize signaling networks in T cells and neuronal glutamate synapses. Results have led to new hypothesis generation with potential diagnostic and therapeutic applications. This protocol includes instructions to perform QMI, from the initial antibody panel selection through to running assays and analyzing data. The initial assembly of a QMI assay involves screening antibodies to generate a panel, and empirically determining an appropriate lysis buffer. The subsequent reagent preparation includes covalently coupling immunoprecipitation antibodies to MagBeads, and biotinylating probe antibodies so they can be labeled by a streptavidin-conjugated fluorophore. To run the assay, lysate is mixed with MagBeads overnight, and then beads are divided and incubated with different probe antibodies, and then a fluorophore label, and read by flow cytometry. Two statistical tests are performed to identify PiSCES that differ significantly between experimental conditions, and results are visualized using heatmaps or node-edge diagrams.
Collapse
Affiliation(s)
- Emily A Brown
- Center for Integrative Brain Research, Seattle Children's Research Institute; Graduate Program in Neuroscience, University of Washington
| | - Steven C Neier
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School; Broad Institute of Harvard and MIT
| | | | - Adam G Schrum
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri; Department of Surgery, School of Medicine, University of Missouri; Department Bioengineering, College of Engineering, University of Missouri
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute; Graduate Program in Neuroscience, University of Washington; Department of Pediatrics, University of Washington;
| |
Collapse
|
18
|
Lautz JD, Gniffke EP, Brown EA, Immendorf KB, Mendel RD, Smith SEP. Activity-dependent changes in synaptic protein complex composition are consistent in different detergents despite differential solubility. Sci Rep 2019; 9:10890. [PMID: 31350430 PMCID: PMC6659712 DOI: 10.1038/s41598-019-46690-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/03/2019] [Indexed: 12/02/2022] Open
Abstract
At the post-synaptic density (PSD), large protein complexes dynamically form and dissociate in response to synaptic activity, comprising the biophysical basis for learning and memory. The use of detergents to isolate the PSD and release its membrane-associated proteins complicates studies of these activity-dependent protein interaction networks, because detergents can simultaneously disrupt the very interactions under study. Despite widespread recognition that different detergents yield different experimental results, the effect of detergent on activity-dependent synaptic protein complexes has not been rigorously examined. Here, we characterize the effect of three detergents commonly used to study synaptic proteins on activity-dependent protein interactions. We first demonstrate that SynGAP-containing interactions are more abundant in 1% Deoxycholate (DOC), while Shank-, Homer- and mGluR5-containing interactions are more abundant in 1% NP-40 or Triton. All interactions were detected preferentially in high molecular weight complexes generated by size exclusion chromatography, although the detergent-specific abundance of proteins in high molecular weight fractions did not correlate with the abundance of detected interactions. Activity-dependent changes in protein complexes were consistent across detergent types, suggesting that detergents do not isolate distinct protein pools with unique behaviors. However, detection of activity-dependent changes is more or less feasible in different detergents due to baseline solubility. Collectively, our results demonstrate that detergents affect the solubility of individual proteins, but activity-dependent changes in protein interactions, when detectable, are consistent across detergent types.
Collapse
Affiliation(s)
- Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Edward P Gniffke
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Emily A Brown
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.,Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| | - Karen B Immendorf
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ryan D Mendel
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA. .,Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA. .,Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
19
|
The genesis and evolution of bead-based multiplexing. Methods 2019; 158:2-11. [DOI: 10.1016/j.ymeth.2019.01.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/10/2018] [Accepted: 01/14/2019] [Indexed: 12/22/2022] Open
|
20
|
Turi KN, Shankar J, Anderson LJ, Rajan D, Gaston K, Gebretsadik T, Das SR, Stone C, Larkin EK, Rosas-Salazar C, Brunwasser SM, Moore ML, Peebles RS, Hartert TV. Infant Viral Respiratory Infection Nasal Immune-Response Patterns and Their Association with Subsequent Childhood Recurrent Wheeze. Am J Respir Crit Care Med 2018; 198:1064-1073. [PMID: 29733679 PMCID: PMC6221572 DOI: 10.1164/rccm.201711-2348oc] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/07/2018] [Indexed: 02/06/2023] Open
Abstract
RATIONALE Recurrent wheeze and asthma are thought to result from alterations in early life immune development following respiratory syncytial virus (RSV) infection. However, prior studies of the nasal immune response to infection have assessed only individual cytokines, which does not capture the whole spectrum of response to infection. OBJECTIVES To identify nasal immune phenotypes in response to RSV infection and their association with recurrent wheeze. METHODS A birth cohort of term healthy infants born June to December were recruited and followed to capture the first infant RSV infection. Nasal wash samples were collected during acute respiratory infection, viruses were identified by RT-PCR, and immune-response analytes were assayed using a multianalyte bead-based panel. Immune-response clusters were identified using machine learning, and association with recurrent wheeze at age 1 and 2 years was assessed using logistic regression. MEASUREMENTS AND MAIN RESULTS We identified two novel and distinct immune-response clusters to RSV and human rhinovirus. In RSV-infected infants, a nasal immune-response cluster characterized by lower non-IFN antiviral immune-response mediators, and higher type-2 and type-17 cytokines was significantly associated with first and second year recurrent wheeze. In comparison, we did not observe this in infants with human rhinovirus acute respiratory infection. Based on network analysis, type-2 and type-17 cytokines were central to the immune response to RSV, whereas growth factors and chemokines were central to the immune response to human rhinovirus. CONCLUSIONS Distinct immune-response clusters during infant RSV infection and their association with risk of recurrent wheeze provide insights into the risk factors for and mechanisms of asthma development.
Collapse
Affiliation(s)
- Kedir N. Turi
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | - Jyoti Shankar
- Infectious Disease Group, J. Craig Venter Institute, Rockville, Maryland; and
| | | | - Devi Rajan
- Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Kelsey Gaston
- Department of Pediatrics, Emory University, Atlanta, Georgia
| | | | - Suman R. Das
- Division of Infectious Diseases, Department of Medicine
- Infectious Disease Group, J. Craig Venter Institute, Rockville, Maryland; and
| | - Cosby Stone
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | - Emma K. Larkin
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | | | | | - Martin L. Moore
- Department of Pediatrics, Emory University, Atlanta, Georgia
| | | | - Tina V. Hartert
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| |
Collapse
|
21
|
Brown EA, Lautz JD, Davis TR, Gniffke EP, VanSchoiack AAW, Neier SC, Tashbook N, Nicolini C, Fahnestock M, Schrum AG, Smith SEP. Clustering the autisms using glutamate synapse protein interaction networks from cortical and hippocampal tissue of seven mouse models. Mol Autism 2018; 9:48. [PMID: 30237867 PMCID: PMC6139139 DOI: 10.1186/s13229-018-0229-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022] Open
Abstract
Background Autism spectrum disorders (ASDs) are a heterogeneous group of behaviorally defined disorders and are associated with hundreds of rare genetic mutations and several environmental risk factors. Mouse models of specific risk factors have been successful in identifying molecular mechanisms associated with a given factor. However, comparisons among different models to elucidate underlying common pathways or to define clusters of biologically relevant disease subtypes have been complicated by different methodological approaches or different brain regions examined by the labs that developed each model. Here, we use a novel proteomic technique, quantitative multiplex co-immunoprecipitation or QMI, to make a series of identical measurements of a synaptic protein interaction network in seven different animal models. We aim to identify molecular disruptions that are common to multiple models. Methods QMI was performed on 92 hippocampal and cortical samples taken from seven mouse models of ASD: Shank3B, Shank3Δex4-9, Ube3a2xTG, TSC2, FMR1, and CNTNAP2 mutants, as well as E12.5 VPA (maternal valproic acid injection on day 12.5 post-conception). The QMI panel targeted a network of 16 interacting, ASD-linked, synaptic proteins, probing 240 potential co-associations. A custom non-parametric statistical test was used to call significant differences between ASD models and littermate controls, and Hierarchical Clustering by Principal Components was used to cluster the models using mean log2 fold change values. Results Each model displayed a unique set of disrupted interactions, but some interactions were disrupted in multiple models. These tended to be interactions that are known to change with synaptic activity. Clustering revealed potential relationships among models and suggested deficits in AKT signaling in Ube3a2xTG mice, which were confirmed by phospho-western blots. Conclusions These data highlight the great heterogeneity among models, but suggest that high-dimensional measures of a synaptic protein network may allow differentiation of subtypes of ASD with shared molecular pathology.
Collapse
Affiliation(s)
- Emily A Brown
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA
| | - Jonathan D Lautz
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA
| | - Tessa R Davis
- 2Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN USA.,3Present address: Department of Biomedical Engineering, UT Austin, Austin, TX USA
| | - Edward P Gniffke
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA
| | - Alison A W VanSchoiack
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA.,Present address: Nanostring, Seattle, WA USA
| | - Steven C Neier
- 2Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN USA.,5Present address: Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA USA.,6Present address: Department of Medicine, Harvard Medical School, Boston, MA USA.,7Present address: Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Noah Tashbook
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA
| | - Chiara Nicolini
- 8Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON Canada
| | - Margaret Fahnestock
- 8Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON Canada
| | - Adam G Schrum
- 9Departments of Molecular Microbiology & Immunology, Surgery and Bioengineering, University of Missouri, Columbia, MO USA
| | - Stephen E P Smith
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA.,10Department of Pediatrics and Graduate Program in Neuroscience, University of Washington, Seattle, WA USA
| |
Collapse
|
22
|
Lautz JD, Brown EA, Williams VanSchoiack AA, Smith SEP. Synaptic activity induces input-specific rearrangements in a targeted synaptic protein interaction network. J Neurochem 2018; 146:540-559. [PMID: 29804286 PMCID: PMC6150823 DOI: 10.1111/jnc.14466] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/04/2018] [Accepted: 05/22/2018] [Indexed: 12/30/2022]
Abstract
Cells utilize dynamic, network-level rearrangements in highly interconnected protein interaction networks to transmit and integrate information from distinct signaling inputs. Despite the importance of protein interaction network dynamics, the organizational logic underlying information flow through these networks is not well understood. Previously, we developed the quantitative multiplex co-immunoprecipitation platform, which allows for the simultaneous and quantitative measurement of the amount of co-association between large numbers of proteins in shared complexes. Here, we adapt quantitative multiplex co-immunoprecipitation to define the activity-dependent dynamics of an 18-member protein interaction network in order to better understand the underlying principles governing glutamatergic signal transduction. We first establish that immunoprecipitation detected by flow cytometry can detect activity-dependent changes in two known protein-protein interactions (Homer1-mGluR5 and PSD-95-SynGAP). We next demonstrate that neuronal stimulation elicits a coordinated change in our targeted protein interaction network, characterized by the initial dissociation of Homer1 and SynGAP-containing complexes followed by increased associations among glutamate receptors and PSD-95. Finally, we show that stimulation of distinct glutamate receptor types results in different modular sets of protein interaction network rearrangements, and that cells activate both modules in order to integrate complex inputs. This analysis demonstrates that cells respond to distinct types of glutamatergic input by modulating different combinations of protein co-associations among a targeted network of proteins. Our data support a model of synaptic plasticity in which synaptic stimulation elicits dissociation of pre-existing multiprotein complexes, opening binding slots in scaffold proteins and allowing for the recruitment of additional glutamatergic receptors. Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Emily A Brown
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, Washington, USA
| |
Collapse
|
23
|
Schrum AG, Neier SC, VanHook AM. Science Signaling Podcast for 2 August 2016: Patient-specific protein complexes. Sci Signal 2016; 9:c17. [PMID: 27485014 DOI: 10.1126/scisignal.aah5912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
This Podcast features an interview with Adam Schrum and Steven Neier, authors of a Research Article that appears in the 2 August 2016 issue of Science Signaling, about a method for identifying protein-protein interactions in patient tissue samples. The authors used this method to compare signaling complexes downstream of the T cell receptor in T cells from healthy skin with those in T cells from the skin of patients with the autoimmune disease alopecia areata. The study revealed differences in the relative abundance of some protein complexes between T cells from the control and patient groups. This technique could be adapted for use as a diagnostic tool to stratify patients by molecular phenotype and predict the therapeutic strategy that is likely to work best for each patient.Listen to Podcast.
Collapse
Affiliation(s)
- Adam G Schrum
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Steven C Neier
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Annalisa M VanHook
- Web Editor, Science Signaling, American Association for the Advancement of Science, 1200 New York Avenue, NW, Washington, DC 20005, USA
| |
Collapse
|