1
|
Lopez-Atalaya JP, Bhojwani-Cabrera AM. Type I interferon signalling and interferon-responsive microglia in health and disease. FEBS J 2025. [PMID: 40299722 DOI: 10.1111/febs.70126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025]
Abstract
Recent evidence suggests that type I interferon (IFN-I) signalling extends beyond its canonical roles in antiviral defence and immunomodulation. Over the past decade, dysregulated IFN-I signalling has been linked to genetic disorders and neurodegenerative diseases, where it may contribute to neurological impairments. Microglia have emerged as key mediators of IFN-I responses in the central nervous system. A distinct transcriptional state responsive to interferons has recently been identified in microglia. The activation of the IFN-I pathway in these cells is now recognised as pivotal in both development and neurodegeneration. This review is divided into two main sections: the first examines the broader role of IFN-I signalling in the central nervous system, particularly its contribution to neurological dysfunction; the second focuses on the specific state of interferon-responsive microglia, exploring its mechanisms and relevance in neurodegenerative conditions. Finally, we discuss how these areas intersect and their implications for both healthy and diseased states.
Collapse
Affiliation(s)
- Jose P Lopez-Atalaya
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Alicante, Spain
| | - Aysha M Bhojwani-Cabrera
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Alicante, Spain
| |
Collapse
|
2
|
Santoro JD, Eduthan NP, Khoshnood MM, Jafarpour S, Boyd NK, Vogel BN, Nguyen L, Kazerooni L, Britton E, Lyford HR, Galbraith MD, Rachubinski AL, Espinosa JM. Evidence of blood-brain barrier dysfunction and CSF immunoglobulin synthesis in Down Syndrome Regression Disorder. Ann Clin Transl Neurol 2025; 12:805-820. [PMID: 39996411 PMCID: PMC12040517 DOI: 10.1002/acn3.52299] [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: 10/24/2024] [Revised: 12/04/2024] [Accepted: 12/29/2024] [Indexed: 02/26/2025] Open
Abstract
OBJECTIVES This study sought to evaluate proteomic, metabolomic, and immune signatures in the cerebrospinal fluid of individuals with Down Syndrome Regression Disorder (DSRD). METHODS A prospective case-control study comparing proteomic, metabolomic, and immune profiles in individuals with DSRD was performed. Samples were obtained from a biorepository of affected individuals and compared to clinically available data and previously obtained neurodiagnostic studies. Individuals with DSRD were compared to individuals with established neuroinflammatory conditions (e.g., multiple sclerosis), and neurotypical controls undergoing a lumbar puncture for headaches. Samples underwent high-throughput proteomic, metabolomic, and immune marker profiling. Data was compared across groups and clinical phenotypes. Gene set enrichment analysis and pathway analyses were utilized to analyze the data. RESULTS In total, 34 individuals with DSRD, 22 neuroinflammatory controls, and 27 neurotypical controls were enrolled in the study. We observed a highly significant concordance in dysregulated proteomics signatures in DSRD and neuroinflammatory controls versus healthy controls, most prominently upregulation of many immunoglobulin sequences. In addition, individuals with DSRD displayed strong upregulation of liver-derived plasma proteins and erythrocyte proteins in the CSF, indicating poor blood-brain barrier integrity. The immune marker profile of DSRD is clearly similar to other neuroimmunological conditions, including strong elevation of MIP3-α, eotaxin, and IFN-γ. INTERPRETATION Individuals with DSRD have unique CSF proteomic and metabolomic signatures consistent with neuroinflammation and increased blood-brain barrier permeability. The CSF of individuals with DSRD was more comparable to individuals with neuroinflammatory disorders than neurotypical controls, indicating the potential for an immune etiology of disease.
Collapse
Affiliation(s)
- Jonathan D. Santoro
- Division of Neurology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
- Department of NeurologyKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Neetha Paul Eduthan
- Linda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Mellad M. Khoshnood
- Division of Neurology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
- Department of NeurologyKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Saba Jafarpour
- Division of Neurology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
- Department of NeurologyKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Natalie K. Boyd
- Division of Neurology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Benjamin N. Vogel
- Division of Neurology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Lina Nguyen
- Division of Neurology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Lilia Kazerooni
- Division of Neurology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Eleanor Britton
- Linda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Hannah R. Lyford
- Linda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Matthew D. Galbraith
- Linda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of PharmacologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Angela L. Rachubinski
- Linda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Division of Developmental Pediatrics, Department of PediatricsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of PharmacologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
3
|
Cozzolino KA, Sanford L, Hunter S, Molison K, Erickson B, Courvan MCS, Jones T, Ajit D, Galbraith MD, Espinosa JM, Bentley D, Allen MA, Dowell RD, Taatjes DJ. Mediator kinase inhibition suppresses hyperactive interferon signaling in Down syndrome. eLife 2025; 13:RP100197. [PMID: 39928031 PMCID: PMC11810109 DOI: 10.7554/elife.100197] [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] [Indexed: 02/11/2025] Open
Abstract
Hyperactive interferon (IFN) signaling is a hallmark of Down syndrome (DS), a condition caused by Trisomy 21 (T21); strategies that normalize IFN signaling could benefit this population. Mediator-associated kinases CDK8 and CDK19 drive inflammatory responses through incompletely understood mechanisms. Using sibling-matched cell lines with/without T21, we investigated Mediator kinase function in the context of hyperactive IFN in DS over a 75 min to 24 hr timeframe. Activation of IFN-response genes was suppressed in cells treated with the CDK8/CDK19 inhibitor cortistatin A (CA), via rapid suppression of IFN-responsive transcription factor (TF) activity. We also discovered that CDK8/CDK19 affect splicing, a novel means by which Mediator kinases control gene expression. To further probe Mediator kinase function, we completed cytokine screens and metabolomics experiments. Cytokines are master regulators of inflammatory responses; by screening 105 different cytokine proteins, we show that Mediator kinases help drive IFN-dependent cytokine responses at least in part through transcriptional regulation of cytokine genes and receptors. Metabolomics revealed that Mediator kinase inhibition altered core metabolic pathways in cell type-specific ways, and broad upregulation of anti-inflammatory lipid mediators occurred specifically in kinase-inhibited cells during hyperactive IFNγ signaling. A subset of these lipids (e.g. oleamide, desmosterol) serve as ligands for nuclear receptors PPAR and LXR, and activation of these receptors occurred specifically during hyperactive IFN signaling in CA-treated cells, revealing mechanistic links between Mediator kinases, lipid metabolism, and nuclear receptor function. Collectively, our results establish CDK8/CDK19 as context-specific metabolic regulators, and reveal that these kinases control gene expression not only via TFs, but also through metabolic changes and splicing. Moreover, we establish that Mediator kinase inhibition antagonizes IFN signaling through transcriptional, metabolic, and cytokine responses, with implications for DS and other chronic inflammatory conditions.
Collapse
Affiliation(s)
- Kira A Cozzolino
- Department of Biochemistry, University of ColoradoBoulderUnited States
| | - Lynn Sanford
- Department of Molecular, Cellular, and Developmental Biology, University of ColoradoBoulderUnited States
- BioFrontiers Institute, University of ColoradoBoulderUnited States
| | - Samuel Hunter
- Department of Molecular, Cellular, and Developmental Biology, University of ColoradoBoulderUnited States
- BioFrontiers Institute, University of ColoradoBoulderUnited States
| | - Kayla Molison
- Department of Biochemistry, University of ColoradoBoulderUnited States
| | - Benjamin Erickson
- Department of Biochemistry and Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
- UC-Denver RNA Bioscience InitiativeAuroraUnited States
| | - Meaghan CS Courvan
- Department of Biochemistry, University of ColoradoBoulderUnited States
- Department of Molecular, Cellular, and Developmental Biology, University of ColoradoBoulderUnited States
- BioFrontiers Institute, University of ColoradoBoulderUnited States
- Crnic Institute Boulder BranchBoulderUnited States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Taylor Jones
- Department of Biochemistry, University of ColoradoBoulderUnited States
| | - Deepa Ajit
- Metabolon Inc, DurhamMorrisvilleUnited States
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pharmacology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Joaquín M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pharmacology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - David Bentley
- Department of Biochemistry and Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
- UC-Denver RNA Bioscience InitiativeAuroraUnited States
| | - Mary Ann Allen
- BioFrontiers Institute, University of ColoradoBoulderUnited States
| | - Robin D Dowell
- Department of Molecular, Cellular, and Developmental Biology, University of ColoradoBoulderUnited States
- BioFrontiers Institute, University of ColoradoBoulderUnited States
| | - Dylan J Taatjes
- Department of Biochemistry, University of ColoradoBoulderUnited States
| |
Collapse
|
4
|
Rachubinski AL, Wallace E, Gurnee E, Enriquez-Estrada BA, Worek KR, Smith KP, Araya P, Waugh KA, Granrath RE, Britton E, Lyford HR, Donovan MG, Eduthan NP, Hill AA, Martin B, Sullivan KD, Patel L, Fidler DJ, Galbraith MD, Dunnick CA, Norris DA, Espinosa JM. JAK inhibition decreases the autoimmune burden in Down syndrome. eLife 2024; 13:RP99323. [PMID: 39737640 DOI: 10.7554/elife.99323] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2025] Open
Abstract
Background Individuals with Down syndrome (DS), the genetic condition caused by trisomy 21 (T21), display clear signs of immune dysregulation, including high rates of autoimmunity and severe complications from infections. Although it is well established that T21 causes increased interferon responses and JAK/STAT signaling, elevated autoantibodies, global immune remodeling, and hypercytokinemia, the interplay between these processes, the clinical manifestations of DS, and potential therapeutic interventions remain ill defined. Methods We report a comprehensive analysis of immune dysregulation at the clinical, cellular, and molecular level in hundreds of individuals with DS, including autoantibody profiling, cytokine analysis, and deep immune mapping. We also report the interim analysis of a Phase II clinical trial investigating the safety and efficacy of the JAK inhibitor tofacitinib through multiple clinical and molecular endpoints. Results We demonstrate multi-organ autoimmunity of pediatric onset concurrent with unexpected autoantibody-phenotype associations in DS. Importantly, constitutive immune remodeling and hypercytokinemia occur from an early age prior to autoimmune diagnoses or autoantibody production. Analysis of the first 10 participants to complete 16 weeks of tofacitinib treatment shows a good safety profile and no serious adverse events. Treatment reduced skin pathology in alopecia areata, psoriasis, and atopic dermatitis, while decreasing interferon scores, cytokine scores, and levels of pathogenic autoantibodies without overt immune suppression. Conclusions JAK inhibition is a valid strategy to treat autoimmune conditions in DS. Additional research is needed to define the effects of JAK inhibition on the broader developmental and clinical hallmarks of DS. Funding NIAMS, Global Down Syndrome Foundation. Clinical trial number NCT04246372.
Collapse
Affiliation(s)
- Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Elizabeth Wallace
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Emily Gurnee
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Belinda A Enriquez-Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Kayleigh R Worek
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Katherine A Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Eleanor Britton
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Hannah R Lyford
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Micah G Donovan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Neetha Paul Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Amanda A Hill
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Barry Martin
- Department of Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Lina Patel
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Psychiatry, Child and Adolescent Division, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Deborah J Fidler
- Department of Human Development and Family Studies, Colorado State University, Fort Collins, United States
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Cory A Dunnick
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - David A Norris
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Joaquín M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, United States
| |
Collapse
|
5
|
Santoro JD, Jafarpour S, Keehan L, Khoshnood MM, Kazerooni L, Boyd NK, Vogel BN, Nguyen L, Manning M, Nagesh D, Spinazzi NA, Besterman AD, Quinn EA, Rafii MS. Diagnostic abnormalities, disease severity and immunotherapy responsiveness in individuals with Down syndrome regression disorder. Sci Rep 2024; 14:30865. [PMID: 39730779 DOI: 10.1038/s41598-024-81819-8] [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: 07/04/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
INTRODUCTION Down Syndrome Regression Disorder (DSRD) is a neuropsychiatric condition causing insomnia, catatonia, encephalopathy, and obsessive-compulsive behavior in otherwise healthy individuals with Down syndrome (DS). Smaller cohorts have identified heterogenous diagnostic abnormalities which have predicted immunotherapy responsiveness although pattern analysis in a large cohort has never been performed. METHODS A multi-center, retrospective study of individuals with DSRD was performed. Individuals met international consensus criteria for DRSD and were aged 10-30 years. Clinical, demographic, and diagnostic data was extracted for all individuals. Serum studies were compared to a group of individuals with DS only. RESULTS A total of 164 individuals with DSRD were identified. Individuals with DSRD were more likely to have a positive antinuclear antibody, low complement 3, abnormal cytokines, and elevated ferritin levels. In a minority of individuals, EEG (30%), MRI (33%) and cerebrospinal fluid (CSF) (21%) were abnormal. Individuals with CSF abnormalities demonstrated greater disease severity at diagnosis on the BFCRS and NPI-Q (p = 0.02 and p < 0.001). Abnormalities in cytokines (p = 0.03), neuroimaging (p < 0.001), and CSF (p = 0.02) were predictive of immunotherapy responsiveness. When MRI and LP were both abnormal or when EEG, MRI and LP were all abnormal, the odds of immunotherapy responsiveness approached 100% (p = 0.01, 95%CI: 1.75-105.1, OR: 13.56 and p = 0.02, 95%CI: 1.37-86.87, OR: 10.91, respectively). CONCLUSIONS In a population of individuals diagnosed with DSRD, abnormalities in serum cytokine levels, neuroimaging findings, and CSF analysis emerged as indicators of disease severity and responsiveness to immunotherapy.
Collapse
Affiliation(s)
- Jonathan D Santoro
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA.
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Saba Jafarpour
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
| | - Laura Keehan
- Department of Pediatrics, Division of Medical Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mellad M Khoshnood
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lilia Kazerooni
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
| | - Natalie K Boyd
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
| | - Benjamin N Vogel
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
| | - Lina Nguyen
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
| | - Melanie Manning
- Department of Pediatrics, Division of Medical Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Deepti Nagesh
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Noemi A Spinazzi
- Benioff Children's Hospital, University of California San Francisco, Oakland, CA, USA
| | - Aaron D Besterman
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Eileen A Quinn
- Department of Pediatrics, University of Toledo, Toledo, OH, USA
| | - Michael S Rafii
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, CA, USA
| |
Collapse
|
6
|
Tallino S, Etebari R, McDonough I, Leon H, Sepulveda I, Winslow W, Bartholomew SK, Perez SE, Mufson EJ, Velazquez R. Assessing the Benefit of Dietary Choline Supplementation Throughout Adulthood in the Ts65Dn Mouse Model of Down Syndrome. Nutrients 2024; 16:4167. [PMID: 39683562 DOI: 10.3390/nu16234167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Down syndrome (DS) is the most common cause of early-onset Alzheimer's disease (AD). Dietary choline has been proposed as a modifiable factor to improve the cognitive and pathological outcomes of AD and DS, especially as many do not reach adequate daily intake levels of choline. While lower circulating choline levels correlate with worse pathological measures in AD patients, choline status and intake in DS is widely understudied. Perinatal choline supplementation (Ch+) in the Ts65Dn mouse model of DS protects offspring against AD-relevant pathology and improves cognition. Further, dietary Ch+ in adult AD models also ameliorates pathology and improves cognition. However, dietary Ch+ in adult Ts65Dn mice has not yet been explored; thus, this study aimed to supply Ch+ throughout adulthood to determine the effects on cognition and DS co-morbidities. METHODS We fed trisomic Ts65Dn mice and disomic littermate controls either a choline normal (ChN; 1.1 g/kg) or a Ch+ (5 g/kg) diet from 4.5 to 14 months of age. RESULTS We found that Ch+ in adulthood failed to improve genotype-specific deficits in spatial learning. However, in both genotypes of female mice, Ch+ significantly improved cognitive flexibility in a reverse place preference task in the IntelliCage behavioral phenotyping system. Further, Ch+ significantly reduced weight gain and peripheral inflammation in female mice of both genotypes, and significantly improved glucose metabolism in male mice of both genotypes. CONCLUSIONS Our findings suggest that adulthood choline supplementation benefits behavioral and biological factors important for general well-being in DS and related to AD risk.
Collapse
Affiliation(s)
- Savannah Tallino
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Rachel Etebari
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Ian McDonough
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Hector Leon
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Isabella Sepulveda
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Wendy Winslow
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Samantha K Bartholomew
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Sylvia E Perez
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Elliott J Mufson
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Ramon Velazquez
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| |
Collapse
|
7
|
Chorvinsky E, Bhattacharya S, Bera BS, Welham A, Ismat K, Lawlor CM, Preciado D, Gomez JL, Morizono H, Pillai DK, Gutierrez MJ, Jaiswal JK, Nino G. Dysregulated airway epithelial antiviral immunity in Down Syndrome impairs type III IFN response and amplifies airway inflammation during RSV infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624921. [PMID: 39651190 PMCID: PMC11623526 DOI: 10.1101/2024.11.22.624921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Trisomy 21 (TS21), also known as Down syndrome (DS), increases pediatric mortality risk from respiratory syncytial virus (RSV) by nine-fold, yet its underlying immunological basis remains unclear. Here, we investigated RSV-induced immunological responses in TS21 airway epithelial cells (AECs), the primary site of respiratory virus entry and host defense. TS21 AECs exhibit hyperactive interferon (IFN) signaling and reduced RSV infectivity, but they also show impaired type-III IFN responses during viral infection. Furthermore, TS21 AECs demonstrate heightened production of proinflammatory mediators CXCL5 and CXCL10 both before and after RSV exposure. Infants with DS suffering from severe viral bronchiolitis demonstrate dysregulated airway immune responses in vivo, characterized by diminished type-III IFN levels and increased CXCL5/CXCL10 secretion. Our results indicate that RSV severity in DS is not due to impaired viral control but to dysregulated airway proinflammatory responses, offering new therapeutic opportunities to mitigate the severity of RSV infection in children with DS.
Collapse
|
8
|
Cozzolino K, Sanford L, Hunter S, Molison K, Erickson B, Courvan MCS, Jones T, Ajit D, Galbraith MD, Espinosa JM, Bentley DL, Allen MA, Dowell RD, Taatjes DJ. Mediator kinase inhibition suppresses hyperactive interferon signaling in Down syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.05.547813. [PMID: 37461585 PMCID: PMC10349994 DOI: 10.1101/2023.07.05.547813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Hyperactive interferon (IFN) signaling is a hallmark of Down syndrome (DS), a condition caused by trisomy 21 (T21); strategies that normalize IFN signaling could benefit this population. Mediator-associated kinases CDK8 and CDK19 drive inflammatory responses through incompletely understood mechanisms. Using sibling-matched cell lines with/without T21, we investigated Mediator kinase function in the context of hyperactive IFN in DS over a 75min - 24h timeframe. Activation of IFN-response genes was suppressed in cells treated with the CDK8/CDK19 inhibitor cortistatin A (CA), via rapid suppression of IFN-responsive transcription factor (TF) activity. We also discovered that CDK8/CDK19 affect splicing, a novel means by which Mediator kinases control gene expression. To further probe Mediator kinase function, we completed cytokine screens and metabolomics experiments. Cytokines are master regulators of inflammatory responses; by screening 105 different cytokine proteins, we show that Mediator kinases help drive IFN-dependent cytokine responses at least in part through transcriptional regulation of cytokine genes and receptors. Metabolomics revealed that Mediator kinase inhibition altered core metabolic pathways in cell type-specific ways, and broad up-regulation of anti-inflammatory lipid mediators occurred specifically in kinase-inhibited cells during hyperactive IFNγ signaling. A subset of these lipids (e.g. oleamide, desmosterol) serve as ligands for nuclear receptors PPAR and LXR, and activation of these receptors occurred specifically during hyperactive IFN signaling in CA-treated cells, revealing mechanistic links between Mediator kinases, lipid metabolism, and nuclear receptor function. Collectively, our results establish CDK8/CDK19 as context-specific metabolic regulators, and reveal that these kinases control gene expression not only via TFs, but also through metabolic changes and splicing. Moreover, we establish that Mediator kinase inhibition antagonizes IFN signaling through transcriptional, metabolic, and cytokine responses, with implications for DS and other chronic inflammatory conditions.
Collapse
Affiliation(s)
- Kira Cozzolino
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
| | - Lynn Sanford
- Dept. of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
| | - Samuel Hunter
- Dept. of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
| | - Kayla Molison
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
| | - Benjamin Erickson
- Dept. Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- UC-Denver RNA Bioscience Initiative
| | - Meaghan C S Courvan
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
- Dept. of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
- Crnic Institute Boulder Branch
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Taylor Jones
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
| | - Deepa Ajit
- Metabolon, Inc., Durham, North Carolina, USA
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Dept. of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Dept. of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David L Bentley
- Dept. Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- UC-Denver RNA Bioscience Initiative
| | - Mary A Allen
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
| | - Robin D Dowell
- Dept. of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
| | - Dylan J Taatjes
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
| |
Collapse
|
9
|
Rakasiwi T, Ryan C, Stein A, Vu A, Dykman M, Shah I, Reilly C, Brokamp G, Mologousis MA, Komishke B, Hou V, Maguiness S, Kirkorian AY, Price H, Hawryluk EB, Fernandez Faith E, Lara-Corrales I, Gurnee E, Holland KE, Rork JF. Dermatologic Conditions in Down Syndrome: A Multi-Site Retrospective Review of International Classification of Diseases Codes. Pediatr Dermatol 2024; 41:1047-1052. [PMID: 39327647 DOI: 10.1111/pde.15757] [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: 03/24/2024] [Revised: 07/27/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND OBJECTIVE As the population and life expectancy of people with Down syndrome increases, identifying common skin conditions throughout the lifespan will help inform clinical care and research. We sought to evaluate dermatologic conditions diagnosed in pediatric and adult patients with Down syndrome. METHODS This multicenter retrospective study analyzed demographic and outpatient visit International Classification of Diseases codes of patients with Down syndrome evaluated at outpatient dermatology clinics in the United States or Canada between 2011 and 2021. RESULTS A total of 1529 patients with Down syndrome were identified from eight academic medical centers: 50.8% were children (0-12 years), 25.2% were adolescents (13-17 years), and 24% were adults (18 years and older). Eczematous dermatitis was the most common diagnosis overall (26%), followed by folliculitis (19.3%) and seborrheic dermatitis (15.6%). Other notable diagnoses included dermatophyte infections (13%), alopecia areata (11.6%), and psoriasis (6.7%). About 4.3% of visits included a code for high-risk medication use. Eczematous dermatitis, alopecia areata, and folliculitis were the most common diagnoses observed in children; folliculitis, hidradenitis suppurativa, and eczematous dermatitis in adolescents; and seborrheic dermatitis, eczematous dermatitis, and folliculitis in adults. CONCLUSIONS Dermatologic conditions in patients with Down syndrome vary by age, but are most often eczematous, adnexal, and cutaneous autoimmune disorders. This multicenter retrospective review identifies skin diseases that should be prioritized for clinical care guideline development and research in the Down syndrome community.
Collapse
Affiliation(s)
- Tasya Rakasiwi
- Department of Dermatology, Dartmouth Health, Manchester, New Hampshire, USA
| | - Chenin Ryan
- Geisel Medical School, Hanover, New Hampshire, USA
| | - Amy Stein
- Pediatric Dermatology Research Alliance, Portland, Oregon, USA
| | - Alan Vu
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Morgan Dykman
- University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Ipsit Shah
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | | | - Bailey Komishke
- Division of Dermatology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Vincent Hou
- University of Colorado Medical School, Aurora, Colorado, USA
| | - Sheilagh Maguiness
- Department of Dermatology, M Health Fairview, Minneapolis, Minnesota, USA
| | - A Yasmine Kirkorian
- Division of Dermatology, Children's National Hospital, Washington, DC, USA
- George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Harper Price
- Division of Dermatology, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Elena B Hawryluk
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Irene Lara-Corrales
- Division of Dermatology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Emily Gurnee
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Kristen E Holland
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jillian F Rork
- Department of Dermatology, Dartmouth Health, Manchester, New Hampshire, USA
- Department of Pediatrics, Dartmouth Health, Manchester, New Hampshire, USA
| |
Collapse
|
10
|
Cuinat S, Bézieau S, Deb W, Mercier S, Vignard V, Isidor B, Küry S, Ebstein F. Understanding neurodevelopmental proteasomopathies as new rare disease entities: A review of current concepts, molecular biomarkers, and perspectives. Genes Dis 2024; 11:101130. [PMID: 39220754 PMCID: PMC11364055 DOI: 10.1016/j.gendis.2023.101130] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/30/2023] [Accepted: 08/19/2023] [Indexed: 09/04/2024] Open
Abstract
The recent advances in high throughput sequencing technology have drastically changed the practice of medical diagnosis, allowing for rapid identification of hundreds of genes causing human diseases. This unprecedented progress has made clear that most forms of intellectual disability that affect more than 3% of individuals worldwide are monogenic diseases. Strikingly, a substantial fraction of the mendelian forms of intellectual disability is associated with genes related to the ubiquitin-proteasome system, a highly conserved pathway made up of approximately 1200 genes involved in the regulation of protein homeostasis. Within this group is currently emerging a new class of neurodevelopmental disorders specifically caused by proteasome pathogenic variants which we propose to designate "neurodevelopmental proteasomopathies". Besides cognitive impairment, these diseases are typically associated with a series of syndromic clinical manifestations, among which facial dysmorphism, motor delay, and failure to thrive are the most prominent ones. While recent efforts have been made to uncover the effects exerted by proteasome variants on cell and tissue landscapes, the molecular pathogenesis of neurodevelopmental proteasomopathies remains ill-defined. In this review, we discuss the cellular changes typically induced by genomic alterations in proteasome genes and explore their relevance as biomarkers for the diagnosis, management, and potential treatment of these new rare disease entities.
Collapse
Affiliation(s)
- Silvestre Cuinat
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Stéphane Bézieau
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Wallid Deb
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Sandra Mercier
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Virginie Vignard
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Bertrand Isidor
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Sébastien Küry
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Frédéric Ebstein
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| |
Collapse
|
11
|
Rachubinski AL, Wallace E, Gurnee E, Estrada BAE, Worek KR, Smith KP, Araya P, Waugh KA, Granrath RE, Britton E, Lyford HR, Donovan MG, Eduthan NP, Hill AA, Martin B, Sullivan KD, Patel L, Fidler DJ, Galbraith MD, Dunnick CA, Norris DA, Espinosa JM. JAK inhibition decreases the autoimmune burden in Down syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.13.24308783. [PMID: 38946973 PMCID: PMC11213071 DOI: 10.1101/2024.06.13.24308783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Individuals with Down syndrome (DS), the genetic condition caused by trisomy 21 (T21), display clear signs of immune dysregulation, including high rates of autoimmune disorders and severe complications from infections. Although it is well established that T21 causes increased interferon responses and JAK/STAT signaling, elevated autoantibodies, global immune remodeling, and hypercytokinemia, the interplay between these processes, the clinical manifestations of DS, and potential therapeutic interventions remain ill defined. Here, we report a comprehensive analysis of immune dysregulation at the clinical, cellular, and molecular level in hundreds of individuals with DS. We demonstrate multi-organ autoimmunity of pediatric onset concurrent with unexpected autoantibody-phenotype associations. Importantly, constitutive immune remodeling and hypercytokinemia occur from an early age prior to autoimmune diagnoses or autoantibody production. We then report the interim analysis of a Phase II clinical trial investigating the safety and efficacy of the JAK inhibitor tofacitinib through multiple clinical and molecular endpoints. Analysis of the first 10 participants to complete the 16-week study shows a good safety profile and no serious adverse events. Treatment reduced skin pathology in alopecia areata, psoriasis, and atopic dermatitis, while decreasing interferon scores, cytokine scores, and levels of pathogenic autoantibodies without overt immune suppression. Additional research is needed to define the effects of JAK inhibition on the broader developmental and clinical hallmarks of DS. ClinicalTrials.gov identifier: NCT04246372.
Collapse
Affiliation(s)
- Angela L. Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth Wallace
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emily Gurnee
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Kayleigh R. Worek
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Keith P. Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katherine A. Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Current address: Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ross E. Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eleanor Britton
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hannah R. Lyford
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Micah G. Donovan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Neetha Paul Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Amanda A. Hill
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Barry Martin
- Department of Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelly D. Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lina Patel
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Psychiatry, Child and Adolescent Division, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Deborah J. Fidler
- Department of Human Development and Family Studies, Colorado State University, Fort Collins, CO, 23 USA
| | - Matthew D. Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cory A. Dunnick
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David A. Norris
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
12
|
Ma X, Li W, Ma J, Han Z, Deng S, Wang S. Autophagy is a promising process for linking inflammation and redox homeostasis in Down syndrome. Front Pharmacol 2024; 15:1491563. [PMID: 39415838 PMCID: PMC11479988 DOI: 10.3389/fphar.2024.1491563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Trisomy 21, characterized by the presence of an additional chromosome 21, leads to a set of clinical features commonly referred to as Down syndrome (DS). The pathological phenotypes observed in DS are caused by a combination of factors, such as mitochondrial dysfunction, neuroinflammation, oxidative stress, disrupted metabolic patterns, and changes in protein homeostasis and signal transduction, and these factors collectively induce neurological alterations. In DS, the triplication of chromosome 21 and the micronuclei arising from the missegregation of chromosomes are closely associated with inflammation and the development of redox imbalance. Autophagy, an essential biological process that affects cellular homeostasis, is a powerful tool to facilitate the degradation of redundant or dysfunctional cytoplasmic components, thereby enabling the recycling of their constituents. Targeting the autophagy process has been suggested as a promising method to balance intracellular inflammation and oxidative stress and improve mitochondrial dysfunction. In this review, we summarize the role of autophagy in regulating inflammation and redox homeostasis in DS and discuss their crosslinks. A comprehensive elucidation of the roles of autophagy in DS offers novel insights for the development of therapeutic strategies aimed at aneuploidy-associated diseases.
Collapse
Affiliation(s)
- Xuehai Ma
- Xinjiang Key Laboratory of Mental Development and Learning Science, College of Psychology, Xinjiang Normal University, Urumqi, Xinjiang, China
| | - Weimin Li
- College of Physical Education and Health, East China Normal University, Shanghai, China
- Institute of Physical Education, Xinjiang Normal University, Urumqi, China
| | - Jun Ma
- Xinjiang Urumqi Youai Hospital, Urumqi, Xinjiang, China
| | - Zhongcheng Han
- People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Shoulong Deng
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Sutian Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
13
|
Rodriguez-Rodriguez C, González-Mancha N, Ochoa-Echeverría A, Mérida I. Sorting nexin 27-dependent regulation of Lck and CD4 tunes the initial stages of T-cell activation. J Leukoc Biol 2024; 116:793-806. [PMID: 38648515 DOI: 10.1093/jleuko/qiae086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Sorting nexin 27 is a unique member of the sorting nexin family of proteins that mediates the endosome-to-plasma membrane trafficking of cargos bearing a PSD95/Dlg1/ZO-1 (PDZ)-binding motif. In brain, sorting nexin 27 regulates synaptic plasticity, and its dysregulation contributes to cognitive impairment and neuronal degeneration. In T lymphocytes, sorting nexin 27 partners with diacylglycerol kinase ζ to facilitate polarized traffic and signaling at the immune synapse. By silencing sorting nexin 27 expression in a human T-cell line, we demonstrate that sorting nexin 27 is a key regulator of the early T-cell tyrosine-based signaling cascade. Sorting nexin 27 transcriptionally controls CD4 abundance in resting conditions and that of its associated molecule, Lck. This guarantees the adequate recruitment of Lck at the immune synapse, which is indispensable for subsequent activation of tyrosine phosphorylation-regulated events. In contrast, reduced sorting nexin 27 expression enhances NF-κB-dependent induction of CXCR4 and triggers production of lytic enzymes and proinflammatory cytokines. These results provide mechanistic explanation to previously described sorting nexin 27 function in the control of immune synapse organization and indicate that impaired sorting nexin 27 expression contributes to CD4 T-cell dysfunction.
Collapse
Affiliation(s)
- Cristina Rodriguez-Rodriguez
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Natalia González-Mancha
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Ane Ochoa-Echeverría
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Isabel Mérida
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
14
|
Donovan MG, Rachubinski AL, Smith KP, Araya P, Waugh KA, Enriquez-Estrada B, Britton EC, Lyford HR, Granrath RE, Schade KA, Kinning KT, Paul Eduthan N, Sullivan KD, Galbraith MD, Espinosa JM. Multimodal analysis of dysregulated heme metabolism, hypoxic signaling, and stress erythropoiesis in Down syndrome. Cell Rep 2024; 43:114599. [PMID: 39120971 PMCID: PMC11479675 DOI: 10.1016/j.celrep.2024.114599] [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: 01/24/2024] [Revised: 06/25/2024] [Accepted: 07/23/2024] [Indexed: 08/11/2024] Open
Abstract
Down syndrome (DS), the genetic condition caused by trisomy 21 (T21), is characterized by delayed neurodevelopment, accelerated aging, and increased risk of many co-occurring conditions. Hypoxemia and dysregulated hematopoiesis have been documented in DS, but the underlying mechanisms and clinical consequences remain ill defined. We report an integrative multi-omic analysis of ∼400 research participants showing that people with DS display transcriptomic signatures indicative of elevated heme metabolism and increased hypoxic signaling across the lifespan, along with chronic overproduction of erythropoietin, elevated biomarkers of tissue-specific hypoxia, and hallmarks of stress erythropoiesis. Elevated heme metabolism, transcriptional signatures of hypoxia, and stress erythropoiesis are conserved in a mouse model of DS and associated with overexpression of select triplicated genes. These alterations are independent of the hyperactive interferon signaling characteristic of DS. These results reveal lifelong dysregulation of key oxygen-related processes that could contribute to the developmental and clinical hallmarks of DS.
Collapse
Affiliation(s)
- Micah G Donovan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katherine A Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Belinda Enriquez-Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eleanor C Britton
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hannah R Lyford
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kyndal A Schade
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kohl T Kinning
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Neetha Paul Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
15
|
Cardiello JF, Westfall J, Dowell R, Allen MA. Characterizing primary transcriptional responses to short term heat shock in Down syndrome. PLoS One 2024; 19:e0307375. [PMID: 39116081 PMCID: PMC11309423 DOI: 10.1371/journal.pone.0307375] [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: 07/06/2023] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Heat shock stress induces genome-wide changes in transcription regulation, activating a coordinated cellular response to enable survival. We noticed many heat shock genes are up-regulated in blood samples from individuals with trisomy 21. We characterized the immediate transcriptional response to heat shock of two lymphoblastoid cell lines derived from brothers with and without trisomy 21. The trisomy 21 cells displayed a more robust heat shock response after just one hour at 42°C than the matched disomic cells.
Collapse
Affiliation(s)
- Joseph F. Cardiello
- BioFrontiers Institute, University of Colorado, Boulder, CO, United States of America
- Department of Chemistry & Biochemistry, Colorado College, Colorado Springs, CO, United States of America
| | - Jessica Westfall
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States of America
| | - Robin Dowell
- BioFrontiers Institute, University of Colorado, Boulder, CO, United States of America
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States of America
- Linda Crnic Institute, University of Colorado, Denver, CO, United States of America
- BioFrontiers-Crnic Boulder Branch, University of Colorado, Boulder, CO, United States of America
| | - Mary Ann Allen
- BioFrontiers Institute, University of Colorado, Boulder, CO, United States of America
- Linda Crnic Institute, University of Colorado, Denver, CO, United States of America
- BioFrontiers-Crnic Boulder Branch, University of Colorado, Boulder, CO, United States of America
| |
Collapse
|
16
|
Rastogi M, Bartolucci M, Nanni M, Aloisio M, Vozzi D, Petretto A, Contestabile A, Cancedda L. Integrative multi-omic analysis reveals conserved cell-projection deficits in human Down syndrome brains. Neuron 2024; 112:2503-2523.e10. [PMID: 38810652 DOI: 10.1016/j.neuron.2024.05.002] [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: 01/02/2023] [Revised: 03/17/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024]
Abstract
Down syndrome (DS) is the most common genetic cause of cognitive disability. However, it is largely unclear how triplication of a small gene subset may impinge on diverse aspects of DS brain physiopathology. Here, we took a multi-omic approach and simultaneously analyzed by RNA-seq and proteomics the expression signatures of two diverse regions of human postmortem DS brains. We found that the overexpression of triplicated genes triggered global expression dysregulation, differentially affecting transcripts, miRNAs, and proteins involved in both known and novel biological candidate pathways. Among the latter, we observed an alteration in RNA splicing, specifically modulating the expression of genes involved in cytoskeleton and axonal dynamics in DS brains. Accordingly, we found an alteration in axonal polarization in neurons from DS human iPSCs and mice. Thus, our study provides an integrated multilayer expression database capable of identifying new potential targets to aid in designing future clinical interventions for DS.
Collapse
Affiliation(s)
- Mohit Rastogi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Marina Nanni
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | | | - Diego Vozzi
- Central RNA Laboratory, Istituto Italiano di Tecnologia, Genova 16152, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy.
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy; Dulbecco Telethon Institute, Rome 00185, Italy.
| |
Collapse
|
17
|
Jafarpour S, Banerjee AK, Khoshnood MM, Vogel BN, Boyd NK, Nguyen L, Partridge R, Santoro SL, Gombolay GY, Fisher KS, de Asua DR, Del Ortega MC, Franklin C, Rafii MS, Santoro JD. De novo variants in immune regulatory genes in Down syndrome regression disorder. J Neurol 2024; 271:5567-5576. [PMID: 38909119 PMCID: PMC11319504 DOI: 10.1007/s00415-024-12521-y] [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: 04/09/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Down Syndrome Regression Disorder (DSRD) is a rare and poorly understood disorder of the central nervous system, characterized by acute or subacute neuropsychiatric symptoms in previously healthy individuals with Down syndrome (DS). Many patients exhibit immunotherapy-responsiveness, indicative of immune dysregulation as a potential underlying etiology. While hypotheses are emerging regarding the role of interferon signaling in DSRD and other autoimmune conditions associated with DS, it is unclear why a small subset of individuals with DS develop DSRD. The aim of this study was to investigate genes of immune regulation in persons with DSRD. METHODS This study included individuals with DSRD aged 10-30 years with trio exome sequencing performed during the diagnostic work up. Descriptive statistics and univariate analysis (Chi-square and Fisher's exact test) were used to describe and compare the characteristics of individuals with and without variants. RESULTS Forty-one individuals with DSRD had trio exome sequencing results. Eight (20%) had heterozygous de novo variants of immune regulatory genes, with four variants being pathogenic or likely pathogenic (UNC13D, XIAP, RNASEH2A, and DNASE1L3). All genes harboring pathogenic variants were associated with interferon type-1 inflammatory response. Individuals harboring variants were more likely to have a preceding trigger (p = 0.03, 95% CI 1.21-97.06), rapid clinical decline in less than 1 month (p = 0.01, 95% CI 1.67-52.06), and MRI abnormalities (p < 0.001, 95% CI 4.89-527.71). DISCUSSION A distinct subset of individuals with DSRD exhibited pathogenic variants in immune regulation genes associated with interferon-mediated inflammatory response, coinciding with previously established links between these genes and interferonopathies such as Aicardi-Goutieres syndrome. Our observations suggest that these variants might potentially contribute to the development of DSRD in individuals with DS.
Collapse
Affiliation(s)
- Saba Jafarpour
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Abhik K Banerjee
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
| | - Mellad M Khoshnood
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
- Los Angeles General Hospital, Los Angeles, CA, USA
| | - Benjamin N Vogel
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
| | - Natalie K Boyd
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
| | - Lina Nguyen
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA
| | | | - Stephanie L Santoro
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Down Syndrome Program, Massachusetts General Hospital, Boston, MA, USA
| | - Grace Y Gombolay
- Division of Neurology, Department of Pediatrics, Emory School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Kristen S Fisher
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Diego Real de Asua
- Adult Down Syndrome Outpatient Clinic, Department of Internal Medicine, Fundación de Investigación Biomédica, Hospital Universitario de La Princesa, Madrid, Spain
| | | | - Cathy Franklin
- Department of Psychiatry, Mater Research Institute, University of Queensland, Brisbane, Australia
| | - Michael S Rafii
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Jonathan D Santoro
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, Mailstop 82, Los Angeles, CA, 90027, USA.
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Donovan MG, Eduthan NP, Smith KP, Britton EC, Lyford HR, Araya P, Granrath RE, Waugh KA, Enriquez Estrada B, Rachubinski AL, Sullivan KD, Galbraith MD, Espinosa JM. Variegated overexpression of chromosome 21 genes reveals molecular and immune subtypes of Down syndrome. Nat Commun 2024; 15:5473. [PMID: 38942750 PMCID: PMC11213896 DOI: 10.1038/s41467-024-49781-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 06/18/2024] [Indexed: 06/30/2024] Open
Abstract
Individuals with Down syndrome, the genetic condition caused by trisomy 21, exhibit strong inter-individual variability in terms of developmental phenotypes and diagnosis of co-occurring conditions. The mechanisms underlying this variable developmental and clinical presentation await elucidation. We report an investigation of human chromosome 21 gene overexpression in hundreds of research participants with Down syndrome, which led to the identification of two major subsets of co-expressed genes. Using clustering analyses, we identified three main molecular subtypes of trisomy 21, based on differential overexpression patterns of chromosome 21 genes. We subsequently performed multiomics comparative analyses among subtypes using whole blood transcriptomes, plasma proteomes and metabolomes, and immune cell profiles. These efforts revealed strong heterogeneity in dysregulation of key pathophysiological processes across the three subtypes, underscored by differential multiomics signatures related to inflammation, immunity, cell growth and proliferation, and metabolism. We also observed distinct patterns of immune cell changes across subtypes. These findings provide insights into the molecular heterogeneity of trisomy 21 and lay the foundation for the development of personalized medicine approaches for the clinical management of Down syndrome.
Collapse
Affiliation(s)
- Micah G Donovan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Neetha P Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Eleanor C Britton
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Hannah R Lyford
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Katherine A Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Belinda Enriquez Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, USA.
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, USA.
| |
Collapse
|
19
|
Hom B, Boyd NK, Vogel BN, Nishimori N, Khoshnood MM, Jafarpour S, Nagesh D, Santoro JD. Down Syndrome and Autoimmune Disease. Clin Rev Allergy Immunol 2024; 66:261-273. [PMID: 38913142 PMCID: PMC11422465 DOI: 10.1007/s12016-024-08996-2] [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] [Accepted: 06/08/2024] [Indexed: 06/25/2024]
Abstract
Down syndrome is the most common genetic cause of intellectual disability and has previously been associated with a variety of autoimmune disorders affecting multiple organ systems. The high prevalence of autoimmune disease, in conjunction with other inflammatory and infectious diseases, in this population suggests an intrinsic immune dysregulation associated with triplication of chromosome 21. Emerging data on the role of chromosome 21 in interferon activation, cytokine production, and activation of B-cell mediated autoimmunity are emerging hypotheses that may explain the elevated prevalence of autoimmune thyroid disease, celiac disease, type I diabetes, autoimmune skin disease, and a variety of autoimmune neurologic conditions. As the life expectancy for individuals with Down syndrome increases, knowledge of the epidemiology, clinical features, management and underlying causes of these conditions will become increasingly important. Disorders such as Hashimoto's thyroiditis are prevalent in between 13 and 34% of individuals with Down syndrome but only 3% of the neurotypical population, a pattern similarly recognized in individuals with Celiac Disease (5.8% v 0.5-2%), alopecia areata (27.7% v. 2%), and vitiligo (4.4% v. 0.05-1.55%), respectively. Given the chronicity of autoimmune conditions, early identification and management can significantly impact the quality of life of individuals with Down syndrome. This comprehensive review will highlight common clinical autoimmune conditions observed in individuals with Down syndrome and explore our current understanding of the mechanisms of disease in this population.
Collapse
Affiliation(s)
- Brian Hom
- Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Natalie K Boyd
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, MS82, Los Angeles, CA90027, USA
| | - Benjamin N Vogel
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, MS82, Los Angeles, CA90027, USA
| | - Nicole Nishimori
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, MS82, Los Angeles, CA90027, USA
| | - Mellad M Khoshnood
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, MS82, Los Angeles, CA90027, USA
| | - Saba Jafarpour
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, MS82, Los Angeles, CA90027, USA
| | - Deepti Nagesh
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, MS82, Los Angeles, CA90027, USA
- Department of Neurology, Keck School of Medicineat the, University of Southern California , Los Angeles, CA, USA
| | - Jonathan D Santoro
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd, MS82, Los Angeles, CA90027, USA.
- Department of Neurology, Keck School of Medicineat the, University of Southern California , Los Angeles, CA, USA.
| |
Collapse
|
20
|
Santoro JD, Jafarpour S, Khoshnood MM, Boyd NK, Vogel BN, Nguyen L, Saucier LE, Partridge R, Tiongson E, Ramos-Platt L, Nagesh D, Ho E, Rosser T, Ahsan N, Mitchell WG, Rafii MS. Safety and tolerability of intravenous immunoglobulin infusion in Down syndrome regression disorder. Am J Med Genet A 2024; 194:e63524. [PMID: 38169137 DOI: 10.1002/ajmg.a.63524] [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: 10/16/2023] [Revised: 12/06/2023] [Accepted: 12/16/2023] [Indexed: 01/05/2024]
Abstract
Three large multi-center studies have identified the clinical utility of intravenous immunoglobulin (IVIg) in the treatment of Down syndrome regression disorder (DSRD). Yet the tolerability of infusions in individuals with DS and the safety of IVIg remains unknown in this population. This study sought to evaluate the safety and tolerability of IVIg in individuals with DSRD compared to a real-world cohort of individuals with pediatric onset neuroimmunologic disorders. A single-center, retrospective chart review evaluating clinically documented infusion reactions was performed for individuals meeting international consensus criteria for DSRD and having IVIg infusions between 2019 and 2023. Infusion reactions were evaluated for severity and need for alterations in infusion plan. This cohort was compared against an age and sex matched cohort of children with neuroimmunologic conditions who had also received IVIg infusions. In total, 127 individuals with DSRD and 186 individuals with other neuroimmunologic disorders were enrolled. There was no difference in the overall rate of adverse reactions (AEs) between the DSRD and general neuroimmunology cohorts (p = 0.31, 95% CI: 0.80-2.00), but cardiac-related AEs specifically were more common among the DSRD group (p = 0.02, 95% CI: 1.23-17.54). When AEs did occur, there was no difference in frequency of pharmacologic intervention (p = 0.12, 95% CI: 0.34-1.13) or discontinuation of therapy (p = 0.74, 95% CI: 0.06-7.44). There was a higher incidence of lab abnormalities on IVIG among the general neuroimmunology cohort (p = 0.03, 95% CI: 0.24-0.94) compared to the DSRD cohort. Transaminitis was the most common laboratory abnormality in the DSRD group. In a large cohort of individuals with DSRD, there were no significant differences in the safety and tolerability of IVIg compared to a cohort of children and young adults with neuroimmunologic conditions.
Collapse
Affiliation(s)
- Jonathan D Santoro
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Saba Jafarpour
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Mellad M Khoshnood
- Department of Pediatrics, Los Angeles General Hospital, Los Angeles, California, USA
| | - Natalie K Boyd
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Benjamin N Vogel
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Lina Nguyen
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Laura E Saucier
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | | | - Emmanuelle Tiongson
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Leigh Ramos-Platt
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Deepti Nagesh
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Eugenia Ho
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Tena Rosser
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Nusrat Ahsan
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Wendy G Mitchell
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Michael S Rafii
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, California, USA
| |
Collapse
|
21
|
Ramacieri G, Locatelli C, Semprini M, Pelleri MC, Caracausi M, Piovesan A, Cicilloni M, Vigna M, Vitale L, Sperti G, Corvaglia LT, Pirazzoli GL, Strippoli P, Catapano F, Vione B, Antonaros F. Zinc metabolism and its role in immunity status in subjects with trisomy 21: chromosomal dosage effect. Front Immunol 2024; 15:1362501. [PMID: 38694501 PMCID: PMC11061464 DOI: 10.3389/fimmu.2024.1362501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/01/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction Trisomy 21 (T21), which causes Down syndrome (DS), is the most common chromosomal aneuploidy in humankind and includes different clinical comorbidities, among which the alteration of the immune system has a heavy impact on patient's lives. A molecule with an important role in immune response is zinc and it is known that its concentration is significantly lower in children with T21. Different hypotheses were made about this metabolic alteration and one of the reasons might be the overexpression of superoxide dismutase 1 (SOD1) gene, as zinc is part of the SOD1 active enzymatic center. Methods The aim of our work is to explore if there is a linear correlation between zinc level and immune cell levels measured in a total of 217 blood samples from subjects with T21. Furthermore, transcriptome map analyses were performed using Transcriptome Mapper (TRAM) software to investigate whether a difference in gene expression is detectable between subjects with T21 and euploid control group in tissues and cells involved in the immune response such as lymphoblastoid cells, thymus and white blood cells. Results Our results have confirmed the literature data stating that the blood zinc level in subjects with T21 is lower compared to the general population; in addition, we report that the T21/control zinc concentration ratio is 2:3, consistent with a chromosomal dosage effect due to the presence of three copies of chromosome 21. The transcriptome map analyses showed an alteration of some gene's expression which might explain low levels of zinc in the blood. Discussion Our data suggest that zinc level is not associated with the levels of immunity cells or proteins analyzed themselves and rather the main role of this ion might be played in altering immune cell function.
Collapse
Affiliation(s)
- Giuseppe Ramacieri
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Specialist School of Child Neuropsychiatry - University of Bologna, Bologna, Italy
| | - Chiara Locatelli
- Neonatology Unit, St. Orsola-Malpighi Polyclinic, Bologna, Italy
| | - Michela Semprini
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Maria Chiara Pelleri
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Maria Caracausi
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Allison Piovesan
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Michela Cicilloni
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Marco Vigna
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Lorenza Vitale
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Giacomo Sperti
- Speciality School of Paediatrics - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Luigi Tommaso Corvaglia
- Neonatology Unit, Department of Medical and Surgical Sciences (DIMEC), St. Orsola-Malpighi Polyclinic, University of Bologna, Bologna, Italy
| | | | - Pierluigi Strippoli
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Francesca Catapano
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Beatrice Vione
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Francesca Antonaros
- Unit of Histology, Embryology and Applied Biology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| |
Collapse
|
22
|
Santoro JD, Khoshnood MM, Jafarpour S, Nguyen L, Boyd NK, Vogel BN, Kammeyer R, Patel L, Manning MA, Rachubinski AL, Filipink RA, Baumer NT, Santoro SL, Franklin C, Tamrazi B, Yeom KW, Worley G, Espinosa JM, Rafii MS. Neuroimaging abnormalities associated with immunotherapy responsiveness in Down syndrome regression disorder. Ann Clin Transl Neurol 2024; 11:1034-1045. [PMID: 38375538 PMCID: PMC11021615 DOI: 10.1002/acn3.52023] [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: 12/19/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
OBJECTIVE To determine the prevalence of neuroimaging abnormalities in individuals with Down syndrome regression disorder (DSRD) and evaluate if neuroimaging abnormalities were predictive of therapeutic responses. METHODS A multicenter, retrospective, case-control study which reviewed neuroimaging studies of individuals with DSRD and compared them to a control cohort of individuals with Down syndrome (DS) alone was performed. Individuals aged 10-30 years and meeting international consensus criteria for DSRD were included. The presence of T1, T2/FLAIR, and SWI signal abnormalities was reviewed. Response rates to various therapies, including immunotherapy, were evaluated in the presence of neuroimaging abnormalities. RESULTS In total, 74 individuals (35%) had either T2/FLAIR and/or SWI signal abnormality compared to 14 individuals (12%) without DSRD (p < 0.001, 95%CI: 2.18-7.63). T2/FLAIR signal abnormalities were not appreciated more frequently in individuals with DSRD (14%, 30/210) than in the control cohort (9%, 11/119) (p = 0.18, OR: 1.63, 95%CI: 0.79-3.40). SWI signal abnormalities were appreciated at a higher frequency in individuals with DSRD (24%, 51/210) compared to the control cohort (4%, 5/119) (p < 0.001, OR: 7.31, 95%CI: 2.83-18.90). T2/FLAIR signal abnormalities were localized to the frontal (40%, 12/30) and parietal lobes (37%, 11/30). SWI signal abnormalities were predominantly in the bilateral basal ganglia (94%, 49/52). Individuals with DSRD and the presence of T2/FLAIR and/or SWI signal abnormalities were much more likely to respond to immunotherapy (p < 0.001, OR: 8.42. 95%CI: 3.78-18.76) and less likely to respond to benzodiazepines (p = 0.01, OR: 0.45, 95%CI: 0.25-0.83), antipsychotics (p < 0.001, OR: 0.28, 95%CI: 0.11-0.55), or electroconvulsive therapy (p < 0.001, OR: 0.12; 95%CI: 0.02-0.78) compared to individuals without these neuroimaging abnormalities. INTERPRETATION This study indicates that in individuals diagnosed with DSRD, T2/FLAIR, and SWI signal abnormalities are more common than previously thought and predict response to immunotherapy.
Collapse
Affiliation(s)
- Jonathan D. Santoro
- Division of Neuroimmunology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
- Department of NeurologyKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Mellad M. Khoshnood
- Division of Neuroimmunology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Saba Jafarpour
- Division of Neuroimmunology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Lina Nguyen
- Division of Neuroimmunology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Natalie K. Boyd
- Division of Neuroimmunology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Benjamin N. Vogel
- Division of Neuroimmunology, Department of PediatricsChildren's Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Ryan Kammeyer
- Department of NeurologyChildren's Hospital ColoradoAuroraColoradoUSA
| | - Lina Patel
- Department of NeurologyChildren's Hospital ColoradoAuroraColoradoUSA
- Department of Pharmacology, Linda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Melanie A. Manning
- Department of GeneticsStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Angela L. Rachubinski
- Department of Pharmacology, Linda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Robyn A. Filipink
- Division of Child Neurology, Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Nicole T. Baumer
- Division of Developmental Medicine, Department of PediatricsBoston Children's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyBoston Children's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Stephanie L. Santoro
- Genetics and Metabolism DivisionMassachusetts General Hospital for ChildrenBostonMassachusettsUSA
- Department of PediatricsHarvard Medical SchoolBostonMassachusettsUSA
| | - Catherine Franklin
- Mater Research Institute‐UQThe University of QueenslandBrisbaneQueenslandAustralia
| | - Benita Tamrazi
- Department of RadiologyChildren's Hospital Los Angeles and Keck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Kristen W. Yeom
- Department of RadiologyStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Gordon Worley
- Department of PediatricsDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Joaquin M. Espinosa
- Department of Pharmacology, Linda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Michael S. Rafii
- Department of NeurologyKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| |
Collapse
|
23
|
Ramba M, Bogunovic D. The immune system in Down Syndrome: Autoimmunity and severe infections. Immunol Rev 2024; 322:300-310. [PMID: 38050836 PMCID: PMC10950520 DOI: 10.1111/imr.13296] [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: 09/01/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 12/07/2023]
Abstract
Over 200,000 individuals in the United States alone live with Down Syndrome (DS), the most common genetic disorder associated with intellectual disability. DS has a constellation of features across the body, including dysregulation of the immune system. Individuals with DS have both a higher frequency of autoimmunity and more severe infections than the general population, highlighting the importance of understanding the immune system in this population. Individuals with DS present with dysregulation of both the innate and adaptive immune systems. Elevated cytokine levels, increased type I and type II IFN signaling, a shift toward memory phenotypes in T cells, and a decrease in the size of the B-cell compartment are observed in individuals with DS, which contribute to both autoinflammation and severe infections. Herein, we discuss the current knowledge of the immune system in individuals with Down Syndrome as well as ideas of necessary further investigations to decipher the mechanisms by which trisomy 21 leads to immune dysregulation, with the ultimate goal of identifying clinical targets to improve treatment.
Collapse
Affiliation(s)
- Meredith Ramba
- Center for Inborn Errors of Immunity, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dusan Bogunovic
- Center for Inborn Errors of Immunity, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
24
|
Hsieh EWY, Bolze A, Hernandez JD. Inborn errors of immunity illuminate mechanisms of human immunology and pave the road to precision medicine. Immunol Rev 2024; 322:5-14. [PMID: 38308392 DOI: 10.1111/imr.13311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Affiliation(s)
- Elena W Y Hsieh
- Department of Pediatrics, Section of Allergy and Immunology, Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, Colorado, USA
| | | | - Joseph D Hernandez
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Stanford University, Stanford, California, USA
| |
Collapse
|
25
|
Pechous RD, Malaviarachchi PA, Xing Z, Douglas A, Crane SD, Theriot HM, Zhang Z, Ghaffarieh A, Huang L, Yu YE, Zhang X. SARS-CoV-2 Infection Causes Heightened Disease Severity and Mortality in a Mouse Model of Down Syndrome. Biomedicines 2024; 12:543. [PMID: 38540156 PMCID: PMC10967796 DOI: 10.3390/biomedicines12030543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 11/03/2024] Open
Abstract
Recent epidemiological studies suggest that individuals with Down syndrome are more susceptible to SARS-CoV-2 infection and have higher rates of hospitalization and mortality than the general population. However, the main drivers behind these disparate health outcomes remain unknown. Herein, we performed experimental infections with SARS-CoV-2 in a well-established mouse model of Down syndrome. We observed similar SARS-CoV-2 replication kinetics and dissemination in the primary and secondary organs between mice with and without Down syndrome, suggesting that both groups have similar susceptibilities to SARS-CoV-2 infection. However, Down syndrome mice exhibited more severe disease as defined by clinical features including symptoms, weight loss, pulmonary function, and survival of mice. We found that increased disease severity in Down syndrome mice could not be attributed solely to increased infectivity or a more dramatic pro-inflammatory response to infection. Rather, results from RNA sequencing suggested that differences in the expression of genes from other physiological pathways, such as deficient oxidative phosphorylation, cardiopulmonary dysfunction, and deficient mucociliary clearance in the lungs may also contribute to heightened disease severity and mortality in Down syndrome mice following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Roger D. Pechous
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Priyangi A. Malaviarachchi
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Zhuo Xing
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Avrium Douglas
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Samantha D. Crane
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Hayley M. Theriot
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Zijing Zhang
- Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alireza Ghaffarieh
- Departments of Ophthalmology and Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Lu Huang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Y. Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Xuming Zhang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
26
|
Rodriguez-Rodriguez C, González-Mancha N, Ochoa-Echeverría A, Liébana R, Merida I. Partial loss of Sorting Nexin 27 resembles age- and Down syndrome-associated T cell dysfunctions. Immun Ageing 2024; 21:2. [PMID: 38166948 PMCID: PMC10759489 DOI: 10.1186/s12979-023-00402-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Sorting Nexin 27 (SNX27)-retromer complex facilitates cargo recycling from endosomes to the plasma membrane. SNX27 downregulation in neurons, as the result of Trisomy 21 (T21), has been linked with cognitive deficits due to impairment of AMPA and NMDA receptor recycling. Studies in human T cell lines likewise demonstrated that SNX27 regulates the correct delivery of cargoes to the immune synapse limiting the activation of pro-inflammatory pathways. Nevertheless, the physiological consequences of partial SNX27 loss in T cell homeostasis are still unclear. RESULTS In this study, we have explored the consequences of T cell specific partial SNX27 downregulation in mice. T cells with partial SNX27 deficiency show a marked deficit in the CD4+ T cell pool, a hallmark of aging in mice and humans, and a well-characterized comorbidity of individuals with Down syndrome (DS). When analyzed ex vivo, CD4+ T cells with partial SNX27 deletion demonstrate enhanced proliferation but diminished IL-2 production. In contrast, the CD8+ population show enhanced expression of pro-inflammatory cytokines and lytic enzymes. CONCLUSIONS This mouse model supports the relevance of SNX27 in the organization of the immune synapse, previously described in cell lines, as well as in the control of T cell homeostasis. Individuals with DS experiment an acceleration of the aging process, which particularly affects the immune and central nervous systems. Thus, we hypothesize that reduced SNX27 expression in DS could contribute to the dysregulation of these systems and further research in SNX27 will shed light on the molecular factors underlying the phenotypes observed in people with DS and its contribution to aging.
Collapse
Affiliation(s)
- Cristina Rodriguez-Rodriguez
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049, Madrid, Spain
| | - Natalia González-Mancha
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049, Madrid, Spain
| | - Ane Ochoa-Echeverría
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049, Madrid, Spain
| | - Rosa Liébana
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049, Madrid, Spain
| | - Isabel Merida
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049, Madrid, Spain.
| |
Collapse
|
27
|
Colvin KL, Elliott RJ, Goodman DM, Harral J, Barrett EG, Yeager ME. Increased lethality of respiratory infection by Streptococcus pneumoniae in the Dp16 mouse model of Down syndrome. FASEB Bioadv 2023; 5:528-540. [PMID: 38094158 PMCID: PMC10714064 DOI: 10.1096/fba.2023-00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/16/2024] Open
Abstract
Objectives We sought to investigate whether the Dp16 mouse model of Down syndrome (DS) is more susceptible to severe and lethal respiratory tract infection by Streptococcus pneumoniae. Study Design We infected controls and Dp16 mice with Streptococcus pneumoniae and measured survival rates. We compared cytokine production by primary lung cell cultures exposed to Streptococcus pneumoniae. We examined lung protein expression for interferon signaling related pathways. We characterized the histopathology and quantified the extent of bronchus-associated lymphoid tissue. Finally, we examined mouse tissues for the presence of oligomeric tau protein. Results We found that the Dp16 mouse model of DS displayed significantly higher susceptibility to lethal respiratory infection with Streptococcus pneumoniae compared to control mice. Lung cells cultured from Dp16 mice displayed unique secreted cytokine profiles compared to control mice. The Dp16 mouse lungs were characterized by profound lobar pneumonia with massive diffuse consolidation involving nearly the entire lobe. Marked red hepatization was noted, and Dp16 mice lungs contained numerous bronchus-associated lymphoid tissues that were highly follicularized. Compared to uninfected mice, both control mice and Dp16 mice infected with Streptococcus pneumoniae showed evidence of oligomeric tau aggregates. Conclusions Increased susceptibility to severe respiratory tract infection with Streptococcus pneumoniae in Dp16 mice closely phenocopies infection in individuals with DS. The increase does not appear to be linked to overexpression of mouse interferon genes syntenic to human chromosome 21.
Collapse
Affiliation(s)
- Kelley L Colvin
- Department of Bioengineering University of Colorado Aurora Colorado USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Aurora Colorado USA
- University of Colorado Denver Health Sciences Aurora Colorado USA
| | - Robert J Elliott
- Department of Bioengineering University of Colorado Aurora Colorado USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Aurora Colorado USA
- University of Colorado Denver Health Sciences Aurora Colorado USA
| | - Desiree M Goodman
- Linda Crnic Institute for Down Syndrome, University of Colorado Aurora Colorado USA
- University of Colorado Denver Health Sciences Aurora Colorado USA
| | - Julie Harral
- University of Colorado Denver Health Sciences Aurora Colorado USA
- Department of Medicine University of Colorado Aurora Colorado USA
| | - Edward G Barrett
- Lovelace Biomedical Research Institute Albuquerque New Mexico USA
| | - Michael E Yeager
- Department of Bioengineering University of Colorado Aurora Colorado USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Aurora Colorado USA
- University of Colorado Denver Health Sciences Aurora Colorado USA
| |
Collapse
|
28
|
Colvin KL, Nguyen K, Boncella KL, Goodman DM, Elliott RJ, Harral JW, Bilodeaux J, Smith BJ, Yeager ME. Lung and Heart Biology of the Dp16 Mouse Model of down Syndrome: Implications for Studying Cardiopulmonary Disease. Genes (Basel) 2023; 14:1819. [PMID: 37761959 PMCID: PMC10530394 DOI: 10.3390/genes14091819] [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: 06/23/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: We sought to investigate the baseline lung and heart biology of the Dp16 mouse model of Down syndrome (DS) as a prelude to the investigation of recurrent respiratory tract infection. (2) Methods: In controls vs. Dp16 mice, we compared peripheral blood cell and plasma analytes. We examined baseline gene expression in lungs and hearts for key parameters related to susceptibility of lung infection. We investigated lung and heart protein expression and performed lung morphometry. Finally, and for the first time each in a model of DS, we performed pulmonary function testing and a hemodynamic assessment of cardiac function. (3) Results: Dp16 mice circulate unique blood plasma cytokines and chemokines. Dp16 mouse lungs over-express the mRNA of triplicated genes, but not necessarily corresponding proteins. We found a sex-specific decrease in the protein expression of interferon α receptors, yet an increased signal transducer and activator of transcription (STAT)-3 and phospho-STAT3. Platelet-activating factor receptor protein was not elevated in Dp16 mice. The lungs of Dp16 mice showed increased stiffness and mean linear intercept and contained bronchus-associated lymphoid tissue. The heart ventricles of Dp16 mice displayed hypotonicity. Finally, Dp16 mice required more ketamine to achieve an anesthetized state. (4) Conclusions: The Dp16 mouse model of DS displays key aspects of lung heart biology akin to people with DS. As such, it has the potential to be an extremely valuable model of recurrent severe respiratory tract infection in DS.
Collapse
Affiliation(s)
- Kelley L. Colvin
- Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO 80045, USA (D.M.G.)
| | - Kathleen Nguyen
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA; (K.N.); (K.L.B.); (R.J.E.); (J.B.); (B.J.S.)
| | - Katie L. Boncella
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA; (K.N.); (K.L.B.); (R.J.E.); (J.B.); (B.J.S.)
| | - Desiree M. Goodman
- Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO 80045, USA (D.M.G.)
| | - Robert J. Elliott
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA; (K.N.); (K.L.B.); (R.J.E.); (J.B.); (B.J.S.)
| | - Julie W. Harral
- Department of Medicine, University of Colorado, Aurora, CO 80045, USA;
| | - Jill Bilodeaux
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA; (K.N.); (K.L.B.); (R.J.E.); (J.B.); (B.J.S.)
| | - Bradford J. Smith
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA; (K.N.); (K.L.B.); (R.J.E.); (J.B.); (B.J.S.)
- Section of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Michael E. Yeager
- Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO 80045, USA (D.M.G.)
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA; (K.N.); (K.L.B.); (R.J.E.); (J.B.); (B.J.S.)
| |
Collapse
|
29
|
Thomas SN, Niemeyer BF, Jimenez-Valdes RJ, Kaiser AJ, Espinosa JM, Sullivan KD, Goodspeed A, Costello JC, Alder JK, Cañas-Arranz R, García-Sastre A, Benam KH. Down syndrome is associated with altered frequency and functioning of tracheal multiciliated cells, and response to influenza virus infection. iScience 2023; 26:107361. [PMID: 37554445 PMCID: PMC10405068 DOI: 10.1016/j.isci.2023.107361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/01/2023] [Accepted: 07/10/2023] [Indexed: 08/10/2023] Open
Abstract
Individuals with Down syndrome (DS) clinically manifest severe respiratory illnesses; however, there is a paucity of data on how DS influences homeostatic physiology of lung airway, and its reactive responses to pulmonary pathogens. We generated well-differentiated ciliated airway epithelia using tracheas from wild-type and Dp(16)1/Yey mice in vitro, and discovered that Dp(16)1/Yey epithelia have significantly lower abundance of ciliated cells, an altered ciliary beating profile, and reduced mucociliary transport. Interestingly, both sets of differentiated epithelia released similar quantities of viral particles after infection with influenza A virus (IAV). However, RNA-sequencing and proteomic analyses revealed an immune hyperreactive phenotype particularly for monocyte-recruiting chemokines in Dp(16)1/Yey epithelia. Importantly, when we challenged mice in vivo with IAV, we observed immune hyper-responsiveness in Dp(16)1/Yey mice, evidenced by higher quantities of lung airway infiltrated monocytes, and elevated levels of pro-inflammatory cytokines in bronchoalveolar lavage fluid. Our findings illuminate mechanisms underlying DS-mediated pathophysiological changes in airway epithelium.
Collapse
Affiliation(s)
- Samantha N. Thomas
- Department of Bioengineering, University of Colorado Denver, Aurora, CO 80045, USA
| | - Brian F. Niemeyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rocio J. Jimenez-Valdes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alexander J. Kaiser
- Department of Bioengineering, University of Colorado Denver, Aurora, CO 80045, USA
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kelly D. Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Andrew Goodspeed
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James C. Costello
- Linda Crnic Institute for Down Syndrome, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jonathan K. Alder
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rodrigo Cañas-Arranz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kambez H. Benam
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
30
|
Santoro JD, Spinazzi NA, Filipink RA, Hayati-Rezvan P, Kammeyer R, Patel L, Sannar EA, Dwyer L, Banerjee AK, Khoshnood M, Jafarpour S, Boyd NK, Partridge R, Gombolay GY, Christy AL, Real de Asua D, Del Carmen Ortega M, Manning MA, Van Mater H, Worley G, Franklin C, Stanley MA, Brown R, Capone GT, Quinn EA, Rafii MS. Immunotherapy responsiveness and risk of relapse in Down syndrome regression disorder. Transl Psychiatry 2023; 13:276. [PMID: 37553347 PMCID: PMC10409776 DOI: 10.1038/s41398-023-02579-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023] Open
Abstract
Down syndrome regression disorder (DSRD) is a clinical symptom cluster consisting of neuropsychiatric regression without an identifiable cause. This study evaluated the clinical effectiveness of IVIg and evaluated clinical characteristics associated with relapse after therapy discontinuation. A prospective, multi-center, non-randomized, observational study was performed. Patients met criteria for DSRD and were treated with IVIg. All patients underwent a standardized wean-off therapy after 9-12 months of treatment. Baseline, on-therapy, and relapse scores of the Neuropsychiatric Inventory Total Score (NPITS), Clinical Global Impression-Severity (CGI-S), and the Bush-Francis Catatonia Rating Scale (BFCRS) were used to track clinical symptoms. Eighty-two individuals were enrolled in this study. Patients had lower BFCRS (MD: -6.68; 95% CI: -8.23, -5.14), CGI-S (MD: -1.27; 95% CI: -1.73, -0.81), and NPITS scores (MD: -6.50; 95% CI: -7.53, -5.47) while they were on therapy compared to baseline. Approximately 46% of the patients (n = 38) experienced neurologic relapse with wean of IVIg. Patients with neurologic relapse were more likely to have any abnormal neurodiagnostic study (χ2 = 11.82, P = 0.001), abnormal MRI (χ2 = 7.78, P = 0.005), and abnormal LP (χ2 = 5.45, P = 0.02), and a personal history of autoimmunity (OR: 6.11, P < 0.001) compared to patients without relapse. IVIg was highly effective in the treatment of DSRD. Individuals with a history of personal autoimmunity or neurodiagnostic abnormalities were more likely to relapse following weaning of immunotherapy, indicating the potential for, a chronic autoimmune etiology in some cases of DSRD.
Collapse
Affiliation(s)
- Jonathan D Santoro
- Division of Neurology, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| | - Noemi A Spinazzi
- Department of Pediatrics, Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Robyn A Filipink
- Division of Child Neurology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Panteha Hayati-Rezvan
- Division of Research on Children, Youth and Families, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Ryan Kammeyer
- Department of Neurology, Children's Hospital of Colorado, Aurora, CO, USA
| | - Lina Patel
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO, USA
| | - Elise A Sannar
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO, USA
| | - Luke Dwyer
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | - Abhik K Banerjee
- Division of Neurology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Mellad Khoshnood
- Division of Neurology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Saba Jafarpour
- Division of Neurology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Natalie K Boyd
- Division of Neurology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | | | - Grace Y Gombolay
- Department of Pediatrics, Division of Neurology Emory University and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | | | - Diego Real de Asua
- Adult Down Syndrome Outpatient Clinic, Department of Internal Medicine, Fundación de Investigación Biomédica, Hospital Universitario de La Princesa, Madrid, Spain
| | | | - Melanie A Manning
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Heather Van Mater
- Division of Rheumatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Gordan Worley
- Division of Pediatric Neurology and Developmental Medicine, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Cathy Franklin
- Queensland Center for Intellectual and Developmental Disability, Mater Research Institute, The University of Queensland, South Brisbane, QLD, Australia
| | - Maria A Stanley
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ruth Brown
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - George T Capone
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Eileen A Quinn
- Department of Pediatrics, University of Toledo College of Medicine and Life Science, Toledo, OH, USA
| | - Michael S Rafii
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
- Alzheimer's Therapeutic Research Institute (ATRI), Keck School of Medicine at the University of Southern California, San Diego, CA, USA
| |
Collapse
|
31
|
Rusu B, Kukreja B, Wu T, Dan SJ, Feng MY, Kalish BT. Single-Nucleus Profiling Identifies Accelerated Oligodendrocyte Precursor Cell Senescence in a Mouse Model of Down Syndrome. eNeuro 2023; 10:ENEURO.0147-23.2023. [PMID: 37491366 PMCID: PMC10449487 DOI: 10.1523/eneuro.0147-23.2023] [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: 05/03/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/27/2023] Open
Abstract
Down syndrome (DS), the most common genetic cause of intellectual disability, is associated with lifelong cognitive deficits. However, the mechanisms by which triplication of chromosome 21 genes drive neuroinflammation and cognitive dysfunction are poorly understood. Here, using the Ts65Dn mouse model of DS, we performed an integrated single-nucleus ATAC and RNA-sequencing (snATAC-seq and snRNA-seq) analysis of the adult cortex. We identified cell type-specific transcriptional and chromatin-associated changes in the Ts65Dn cortex, including regulators of neuroinflammation, transcription and translation, myelination, and mitochondrial function. We discovered enrichment of a senescence-associated transcriptional signature in Ts65Dn oligodendrocyte (OL) precursor cells (OPCs) and epigenetic changes consistent with a loss of heterochromatin. We found that senescence is restricted to a subset of OPCs concentrated in deep cortical layers. Treatment of Ts65Dn mice with a senescence-reducing flavonoid rescued cortical OPC proliferation, restored microglial homeostasis, and improved contextual fear memory. Together, these findings suggest that cortical OPC senescence may be an important driver of neuropathology in DS.
Collapse
Affiliation(s)
- Bianca Rusu
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Bharti Kukreja
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Taiyi Wu
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Sophie J Dan
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Min Yi Feng
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Brian T Kalish
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Division of Neonatology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
32
|
Chi C, Knight WE, Riching AS, Zhang Z, Tatavosian R, Zhuang Y, Moldovan R, Rachubinski AL, Gao D, Xu H, Espinosa JM, Song K. Interferon hyperactivity impairs cardiogenesis in Down syndrome via downregulation of canonical Wnt signaling. iScience 2023; 26:107012. [PMID: 37360690 PMCID: PMC10285545 DOI: 10.1016/j.isci.2023.107012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/03/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
Congenital heart defects (CHDs) are frequent in children with Down syndrome (DS), caused by trisomy of chromosome 21. However, the underlying mechanisms are poorly understood. Here, using a human-induced pluripotent stem cell (iPSC)-based model and the Dp(16)1Yey/+ (Dp16) mouse model of DS, we identified downregulation of canonical Wnt signaling downstream of increased dosage of interferon (IFN) receptors (IFNRs) genes on chromosome 21 as a causative factor of cardiogenic dysregulation in DS. We differentiated human iPSCs derived from individuals with DS and CHDs, and healthy euploid controls into cardiac cells. We observed that T21 upregulates IFN signaling, downregulates the canonical WNT pathway, and impairs cardiac differentiation. Furthermore, genetic and pharmacological normalization of IFN signaling restored canonical WNT signaling and rescued defects in cardiogenesis in DS in vitro and in vivo. Our findings provide insights into mechanisms underlying abnormal cardiogenesis in DS, ultimately aiding the development of therapeutic strategies.
Collapse
Affiliation(s)
- Congwu Chi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Walter E. Knight
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Andrew S. Riching
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Zhen Zhang
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Roubina Tatavosian
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Yonghua Zhuang
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Radu Moldovan
- Department of Pharmacology, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Angela L. Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Dexiang Gao
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Hongyan Xu
- Department of Population Health Sciences, Medical College of Georgia, Augusta University; Augusta, GA 30912, USA
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus; Aurora, CO 80045, USA
| |
Collapse
|
33
|
Janoff EN, Tseng HF, Nguyen JL, Alfred T, Vietri J, McDaniel A, Chilson E, Yan Q, Malhotra D, Isturiz RE, Levin MJ. Incidence and clinical outcomes of pneumonia in persons with down syndrome in the United States. Vaccine 2023; 41:4571-4578. [PMID: 37328350 DOI: 10.1016/j.vaccine.2023.05.063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/04/2023] [Accepted: 05/26/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Persons with Down syndrome (DS) experience an increased risk of pneumonia. We determined the incidence and outcomes of pneumonia and relationship to underlying comorbidities in persons with and without DS in the United States. METHODS This retrospective matched cohort study used de-identified administrative claims data from Optum. Persons with DS were matched 1:4 to persons without DS on age, sex, and race/ethnicity. Pneumonia episodes were analyzed for incidence, rate ratios and 95 % confidence intervals, clinical outcomes, and comorbidities. RESULTS During 1-year follow-up among 33796 persons with and 135184 without DS, the incidence of all-cause pneumonia (pneumonia) was substantially higher among people with DS than those without DS (12427 vs. 2531 episodes/100000 person-years; 4.7-5.7 fold increase). Persons with DS and pneumonia were more likely to be hospitalized (39.4 % vs. 13.9 %) or admitted to the ICU (16.8 % vs. 4.8 %). Mortality was higher 1 year after first pneumonia (5.7 % vs. 2.4 %; P < 0.0001). Results were similar for episodes of pneumococcal pneumonia. Specific comorbidities were associated with pneumonia, particularly heart disease in children and neurologic disease in adults, which only partially mediated the effect of DS on pneumonia. CONCLUSIONS Among persons with DS, incidence of pneumonia and associated hospitalizations were increased; mortality among those with pneumonia was comparable at 30 days, but higher at 1 year. DS should be considered an independent risk condition for pneumonia.
Collapse
Affiliation(s)
- Edward N Janoff
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA; Denver Veterans Affairs Medical Center, Aurora, CO, USA.
| | - Hung-Fu Tseng
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Jennifer L Nguyen
- Medical Development and Scientific/Clinical Affairs, Pfizer Vaccines, Collegeville, PA, USA
| | - Tamuno Alfred
- Statistical Research and Data Science Center, Pfizer Inc, New York, NY, USA
| | - Jeffrey Vietri
- Patient and Health Impact, Pfizer Inc, Collegeville, PA, USA
| | - Angee McDaniel
- Medical Development and Scientific/Clinical Affairs, Pfizer Vaccines, Collegeville, PA, USA
| | - Erica Chilson
- Medical Development and Scientific/Clinical Affairs, Pfizer Vaccines, Collegeville, PA, USA
| | - Qi Yan
- Medical Development and Scientific/Clinical Affairs, Pfizer Vaccines, Collegeville, PA, USA
| | - Deepa Malhotra
- Patient and Health Impact, Pfizer Inc, New York, NY, USA
| | - Raul E Isturiz
- Medical Development and Scientific/Clinical Affairs, Pfizer Vaccines, Collegeville, PA, USA
| | - Myron J Levin
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA; Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
34
|
Elling CL, Goff SH, Hirsch SD, Tholen K, Kofonow JM, Curtis D, Robertson CE, Prager JD, Yoon PJ, Wine TM, Chan KH, Scholes MA, Friedman NR, Frank DN, Herrmann BW, Santos-Cortez RLP. Otitis Media in Children with Down Syndrome Is Associated with Shifts in the Nasopharyngeal and Middle Ear Microbiotas. Genet Test Mol Biomarkers 2023; 27:221-228. [PMID: 37522794 PMCID: PMC10494904 DOI: 10.1089/gtmb.2023.0132] [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] [Indexed: 08/01/2023] Open
Abstract
Background: Otitis media (OM) is defined as middle ear (ME) inflammation that is usually due to infection. Globally, OM is a leading cause of hearing loss and is the most frequently diagnosed disease in young children. For OM, pediatric patients with Down syndrome (DS) demonstrate higher incidence rates, greater severity, and poorer outcomes. However, to date, no studies have investigated the bacterial profiles of children with DS and OM. Method: We aimed to determine if there are differences in composition of bacterial profiles or the relative abundance of individual taxa within the ME and nasopharyngeal (NP) microbiotas of pediatric OM patients with DS (n = 11) compared with those without DS (n = 84). We sequenced the 16S rRNA genes and analyzed the sequence data for diversity indices and relative abundance of individual taxa. Results: Individuals with DS demonstrated increased biodiversity in their ME and NP microbiotas. In children with OM, DS was associated with increased biodiversity and higher relative abundance of specific taxa in the ME. Conclusion: Our findings suggest that dysbioses in the NP of DS children contributes to their increased susceptibility to OM compared with controls. These findings suggest that DS influences regulation of the mucosal microbiota and contributes to OM pathology.
Collapse
Affiliation(s)
- Christina L. Elling
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Salina H. Goff
- Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Scott D. Hirsch
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kaitlyn Tholen
- Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Jennifer M. Kofonow
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Danielle Curtis
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Charles E. Robertson
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jeremy D. Prager
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Patricia J. Yoon
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Todd M. Wine
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Kenny H. Chan
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Melissa A. Scholes
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Norman R. Friedman
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brian W. Herrmann
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Regie Lyn P. Santos-Cortez
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Center for Children's Surgery, Children's Hospital Colorado, Aurora, Colorado, USA
| |
Collapse
|
35
|
Galbraith MD, Rachubinski AL, Smith KP, Araya P, Waugh KA, Enriquez-Estrada B, Worek K, Granrath RE, Kinning KT, Paul Eduthan N, Ludwig MP, Hsieh EW, Sullivan KD, Espinosa JM. Multidimensional definition of the interferonopathy of Down syndrome and its response to JAK inhibition. SCIENCE ADVANCES 2023; 9:eadg6218. [PMID: 37379383 PMCID: PMC10306300 DOI: 10.1126/sciadv.adg6218] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023]
Abstract
Individuals with Down syndrome (DS) display chronic hyperactivation of interferon signaling. However, the clinical impacts of interferon hyperactivity in DS are ill-defined. Here, we describe a multiomics investigation of interferon signaling in hundreds of individuals with DS. Using interferon scores derived from the whole blood transcriptome, we defined the proteomic, immune, metabolic, and clinical features associated with interferon hyperactivity in DS. Interferon hyperactivity associates with a distinct proinflammatory phenotype and dysregulation of major growth signaling and morphogenic pathways. Individuals with the highest interferon activity display the strongest remodeling of the peripheral immune system, including increased cytotoxic T cells, B cell depletion, and monocyte activation. Interferon hyperactivity accompanies key metabolic changes, most prominently dysregulated tryptophan catabolism. High interferon signaling stratifies a subpopulation with elevated rates of congenital heart disease and autoimmunity. Last, a longitudinal case study demonstrated that JAK inhibition normalizes interferon signatures with therapeutic benefit in DS. Together, these results justify the testing of immune-modulatory therapies in DS.
Collapse
Affiliation(s)
- Matthew D. Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angela L. Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Keith P. Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katherine A. Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Belinda Enriquez-Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kayleigh Worek
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross E. Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kohl T. Kinning
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Neetha Paul Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael P. Ludwig
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elena W. Y. Hsieh
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Division of Allergy/Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelly D. Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
36
|
Genes with immune functions modulate key traits associated with Down syndrome in mice. Nat Genet 2023:10.1038/s41588-023-01400-3. [PMID: 37277651 DOI: 10.1038/s41588-023-01400-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
|
37
|
Waugh KA, Minter R, Baxter J, Chi C, Galbraith MD, Tuttle KD, Eduthan NP, Kinning KT, Andrysik Z, Araya P, Dougherty H, Dunn LN, Ludwig M, Schade KA, Tracy D, Smith KP, Granrath RE, Busquet N, Khanal S, Anderson RD, Cox LL, Estrada BE, Rachubinski AL, Lyford HR, Britton EC, Fantauzzo KA, Orlicky DJ, Matsuda JL, Song K, Cox TC, Sullivan KD, Espinosa JM. Triplication of the interferon receptor locus contributes to hallmarks of Down syndrome in a mouse model. Nat Genet 2023; 55:1034-1047. [PMID: 37277650 PMCID: PMC10260402 DOI: 10.1038/s41588-023-01399-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 04/14/2023] [Indexed: 06/07/2023]
Abstract
Down syndrome (DS), the genetic condition caused by trisomy 21, is characterized by variable cognitive impairment, immune dysregulation, dysmorphogenesis and increased prevalence of diverse co-occurring conditions. The mechanisms by which trisomy 21 causes these effects remain largely unknown. We demonstrate that triplication of the interferon receptor (IFNR) gene cluster on chromosome 21 is necessary for multiple phenotypes in a mouse model of DS. Whole-blood transcriptome analysis demonstrated that IFNR overexpression associates with chronic interferon hyperactivity and inflammation in people with DS. To define the contribution of this locus to DS phenotypes, we used genome editing to correct its copy number in a mouse model of DS, which normalized antiviral responses, prevented heart malformations, ameliorated developmental delays, improved cognition and attenuated craniofacial anomalies. Triplication of the Ifnr locus modulates hallmarks of DS in mice, suggesting that trisomy 21 elicits an interferonopathy potentially amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Katherine A Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross Minter
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jessica Baxter
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Congwu Chi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kathryn D Tuttle
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Neetha P Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kohl T Kinning
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zdenek Andrysik
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hannah Dougherty
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lauren N Dunn
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael Ludwig
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kyndal A Schade
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dayna Tracy
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nicolas Busquet
- Animal Behavior Core, NeuroTechnology Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Santosh Khanal
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ryan D Anderson
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Liza L Cox
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Belinda Enriquez Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hannah R Lyford
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eleanor C Britton
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katherine A Fantauzzo
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David J Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer L Matsuda
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | - Kunhua Song
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Timothy C Cox
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
38
|
Ebstein F, Küry S, Most V, Rosenfelt C, Scott-Boyer MP, van Woerden GM, Besnard T, Papendorf JJ, Studencka-Turski M, Wang T, Hsieh TC, Golnik R, Baldridge D, Forster C, de Konink C, Teurlings SM, Vignard V, van Jaarsveld RH, Ades L, Cogné B, Mignot C, Deb W, Jongmans MC, Sessions Cole F, van den Boogaard MJH, Wambach JA, Wegner DJ, Yang S, Hannig V, Brault JA, Zadeh N, Bennetts B, Keren B, Gélineau AC, Powis Z, Towne M, Bachman K, Seeley A, Beck AE, Morrison J, Westman R, Averill K, Brunet T, Haasters J, Carter MT, Osmond M, Wheeler PG, Forzano F, Mohammed S, Trakadis Y, Accogli A, Harrison R, Guo Y, Hakonarson H, Rondeau S, Baujat G, Barcia G, Feichtinger RG, Mayr JA, Preisel M, Laumonnier F, Kallinich T, Knaus A, Isidor B, Krawitz P, Völker U, Hammer E, Droit A, Eichler EE, Elgersma Y, Hildebrand PW, Bolduc F, Krüger E, Bézieau S. PSMC3 proteasome subunit variants are associated with neurodevelopmental delay and type I interferon production. Sci Transl Med 2023; 15:eabo3189. [PMID: 37256937 PMCID: PMC10506367 DOI: 10.1126/scitranslmed.abo3189] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/10/2023] [Indexed: 06/02/2023]
Abstract
A critical step in preserving protein homeostasis is the recognition, binding, unfolding, and translocation of protein substrates by six AAA-ATPase proteasome subunits (ATPase-associated with various cellular activities) termed PSMC1-6, which are required for degradation of proteins by 26S proteasomes. Here, we identified 15 de novo missense variants in the PSMC3 gene encoding the AAA-ATPase proteasome subunit PSMC3/Rpt5 in 23 unrelated heterozygous patients with an autosomal dominant form of neurodevelopmental delay and intellectual disability. Expression of PSMC3 variants in mouse neuronal cultures led to altered dendrite development, and deletion of the PSMC3 fly ortholog Rpt5 impaired reversal learning capabilities in fruit flies. Structural modeling as well as proteomic and transcriptomic analyses of T cells derived from patients with PSMC3 variants implicated the PSMC3 variants in proteasome dysfunction through disruption of substrate translocation, induction of proteotoxic stress, and alterations in proteins controlling developmental and innate immune programs. The proteostatic perturbations in T cells from patients with PSMC3 variants correlated with a dysregulation in type I interferon (IFN) signaling in these T cells, which could be blocked by inhibition of the intracellular stress sensor protein kinase R (PKR). These results suggest that proteotoxic stress activated PKR in patient-derived T cells, resulting in a type I IFN response. The potential relationship among proteosome dysfunction, type I IFN production, and neurodevelopment suggests new directions in our understanding of pathogenesis in some neurodevelopmental disorders.
Collapse
Affiliation(s)
- Frédéric Ebstein
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Sébastien Küry
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, 44000 Nantes, France
| | - Victoria Most
- Institut für Medizinische Physik und Biophysik, Universität Leipzig, Medizinische Fakultät, Härtelstr. 16-18, 04107 Leipzig, Germany
| | - Cory Rosenfelt
- Department of Pediatrics, University of Alberta, Edmonton, AB CT6G 1C9, Canada
| | | | - Geeske M. van Woerden
- Department of Neuroscience, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Thomas Besnard
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, 44000 Nantes, France
| | - Jonas Johannes Papendorf
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Maja Studencka-Turski
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Tianyun Wang
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
- Neuroscience Research Institute, Peking University; Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing 100191, China
| | - Tzung-Chien Hsieh
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Richard Golnik
- Klinik für Pädiatrie I, Universitätsklinikum Halle (Saale), 06120 Halle (Saale)
| | - Dustin Baldridge
- The Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63130-4899, USA
| | - Cara Forster
- GeneDx, 207 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Charlotte de Konink
- Department of Neuroscience, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Selina M.W. Teurlings
- Department of Neuroscience, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Virginie Vignard
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, 44000 Nantes, France
| | | | - Lesley Ades
- Department of Clinical Genetics, The Children’s Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
- Disciplines of Genomic Medicine & Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2145, Australia
| | - Benjamin Cogné
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, 44000 Nantes, France
| | - Cyril Mignot
- APHP, Hôpital Pitié-Salpêtrière, Département de Génétique, Centre de Reference Déficience Intellectuelle de Causes Rares, GRC UPMC «Déficience Intellectuelle et Autisme», 75013 Paris, France
- Sorbonne Universités, Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR 7225, 75013, Paris, France
| | - Wallid Deb
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, 44000 Nantes, France
| | - Marjolijn C.J. Jongmans
- Department of Genetics, University Medical Center Utrecht, 3508 AB, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | - F. Sessions Cole
- The Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63130-4899, USA
| | | | - Jennifer A. Wambach
- The Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63130-4899, USA
| | - Daniel J. Wegner
- The Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63130-4899, USA
| | - Sandra Yang
- GeneDx, 207 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Vickie Hannig
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jennifer Ann Brault
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Neda Zadeh
- Genetics Center, Orange, CA 92868, USA; Division of Medical Genetics, Children’s Hospital of Orange County, Orange, CA 92868, USA
| | - Bruce Bennetts
- Disciplines of Genomic Medicine & Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2145, Australia
- Sydney Genome Diagnostics, Western Sydney Genetics Program, The Children’s Hospital at Westmead, Sydney, NSW, 2145, Australia
| | - Boris Keren
- Département de Génétique, Centre de Référence des Déficiences Intellectuelles de Causes Rares, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, 75013 Paris
| | - Anne-Claire Gélineau
- Département de Génétique, Centre de Référence des Déficiences Intellectuelles de Causes Rares, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, 75013 Paris
| | - Zöe Powis
- Department of Clinical Research, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Meghan Towne
- Department of Clinical Research, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | | | - Andrea Seeley
- Genomic Medicine Institute, Geisinger, Danville, PA 17822, USA
| | - Anita E. Beck
- Department of Pediatrics, Division of Genetic Medicine, University of Washington & Seattle Children’s Hospital, Seattle, WA 98195-6320, USA
| | - Jennifer Morrison
- Division of Genetics, Arnold Palmer Hospital for Children, Orlando Health, Orlando, FL 32806, USA
| | - Rachel Westman
- Division of Genetics, St. Luke’s Clinic, Boise, ID 83712, USA
| | - Kelly Averill
- Department of Pediatrics, Division of Pediatric Neurology, UT Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Theresa Brunet
- Institute of Human Genetics, Technical University of Munich, School of Medicine, 81675 Munich, Germany
- Institute of Neurogenomics (ING), Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Judith Haasters
- Klinikum der Universität München, Integriertes Sozial- pädiatrisches Zentrum, 80337 Munich, Germany
| | - Melissa T. Carter
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, ON K1H 8L1, Canada
- Department of Genetics, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - Matthew Osmond
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, ON K1H 8L1, Canada
| | - Patricia G. Wheeler
- Division of Genetics, Arnold Palmer Hospital for Children, Orlando Health, Orlando, FL 32806, USA
| | - Francesca Forzano
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Clinical Genetics Department, Guy’s & St Thomas’ NHS Foundation Trust, London SE1 9RT, UK
| | - Shehla Mohammed
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Clinical Genetics Department, Guy’s & St Thomas’ NHS Foundation Trust, London SE1 9RT, UK
| | - Yannis Trakadis
- Division of Medical Genetics, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Andrea Accogli
- Division of Medical Genetics, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Rachel Harrison
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Department of Clinical Genetics, Nottingham University Hospitals NHS Trust, City Hospital Campus, The Gables, Gate 3, Hucknall Road, Nottingham NG5 1PB, UK
| | - Yiran Guo
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Center for Data Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sophie Rondeau
- Service de Médecine Génomique des Maladies Rares, Hôpital Universitaire Necker-Enfants Malades, 75743 Paris, France
| | - Geneviève Baujat
- Service de Médecine Génomique des Maladies Rares, Hôpital Universitaire Necker-Enfants Malades, 75743 Paris, France
| | - Giulia Barcia
- Service de Médecine Génomique des Maladies Rares, Hôpital Universitaire Necker-Enfants Malades, 75743 Paris, France
| | - René Günther Feichtinger
- University Children’s Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Johannes Adalbert Mayr
- University Children’s Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Martin Preisel
- University Children’s Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Frédéric Laumonnier
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
- Service de Génétique, Centre Hospitalier Régional Universitaire, 37032 Tours, France
| | - Tilmann Kallinich
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin; 13353 Berlin, Germany
- Deutsches Rheumaforschungszentrum, An Institute of the Leibniz Association, Berlin and Berlin Institute of Health, 10117 Berlin, Germany
| | - Alexej Knaus
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Bertrand Isidor
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, 44000 Nantes, France
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Uwe Völker
- Universitätsmedizin Greifswald, Interfakultäres Institut für Genetik und Funktionelle Genomforschung, Abteilung für Funktionelle Genomforschung, 17487 Greifswald, Germany
| | - Elke Hammer
- Universitätsmedizin Greifswald, Interfakultäres Institut für Genetik und Funktionelle Genomforschung, Abteilung für Funktionelle Genomforschung, 17487 Greifswald, Germany
| | - Arnaud Droit
- Research Center of Quebec CHU-Université Laval, Québec, QC PQ G1E6W2, Canada
| | - Evan E. Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Ype Elgersma
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Peter W. Hildebrand
- Institut für Medizinische Physik und Biophysik, Universität Leipzig, Medizinische Fakultät, Härtelstr. 16-18, 04107 Leipzig, Germany
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Berlin, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
| | - François Bolduc
- Department of Pediatrics, University of Alberta, Edmonton, AB CT6G 1C9, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Elke Krüger
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Stéphane Bézieau
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, 44000 Nantes, France
| |
Collapse
|
39
|
Tielemans B, De Herdt L, Pollenus E, Vanhulle E, Seldeslachts L, Marain F, Belmans F, Ahookhosh K, Vanoirbeek J, Vermeire K, Van den Steen PE, Vande Velde G. A Multimodal Imaging-Supported Down Syndrome Mouse Model of RSV Infection. Viruses 2023; 15:v15040993. [PMID: 37112973 PMCID: PMC10144178 DOI: 10.3390/v15040993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Individuals with Down syndrome (DS) are more prone to develop severe respiratory tract infections. Although a RSV infection has a high clinical impact and severe outcome in individuals with DS, no vaccine nor effective therapeutics are available. Any research into infection pathophysiology or prophylactic and therapeutic antiviral strategies in the specific context of DS would greatly benefit this patient population, but currently such relevant animal models are lacking. This study aimed to develop and characterize the first mouse model of RSV infection in a DS-specific context. Ts65Dn mice and wild type littermates were inoculated with a bioluminescence imaging-enabled recombinant human RSV to longitudinally track viral replication in host cells throughout infection progression. This resulted in an active infection in the upper airways and lungs with similar viral load in Ts65Dn mice and euploid mice. Flow cytometric analysis of leukocytes in lungs and spleen demonstrated immune alterations with lower CD8+ T cells and B-cells in Ts65Dn mice. Overall, our study presents a novel DS-specific mouse model of hRSV infection and shows that potential in using the Ts65Dn preclinical model to study immune-specific responses of RSV in the context of DS and supports the need for models representing the pathological development.
Collapse
Affiliation(s)
- Birger Tielemans
- Biomedical MRI Unit/Mosaic, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Lander De Herdt
- Biomedical MRI Unit/Mosaic, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Emiel Vanhulle
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Laura Seldeslachts
- Biomedical MRI Unit/Mosaic, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Fopke Marain
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Flore Belmans
- Biomedical MRI Unit/Mosaic, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
- Radiomics (Oncoradiomics SA), 4000 Liege, Belgium
| | - Kaveh Ahookhosh
- Biomedical MRI Unit/Mosaic, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Jeroen Vanoirbeek
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium
| | - Kurt Vermeire
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Philippe E Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Greetje Vande Velde
- Biomedical MRI Unit/Mosaic, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
40
|
Hamlett ED, Flores-Aguilar L, Handen B, Potier MC, Granholm AC, Sherman S, Puig V, Santoro JD, Carmona-Iragui M, Rebillat AS, Head E, Strydom A, Busciglio J. Innovating Therapies for Down Syndrome: An International Virtual Conference of the T21 Research Society. Mol Syndromol 2023; 14:89-100. [PMID: 37064334 PMCID: PMC10090974 DOI: 10.1159/000526021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Research focused on Down syndrome continued to gain momentum in the last several years and is advancing our understanding of how trisomy 21 (T21) modifies molecular and cellular processes. The Trisomy 21 Research Society (T21RS) is the premier scientific organization for researchers and clinicians studying Down syndrome. During the COVID pandemic, T21RS held its first virtual conference program, sponsored by the University of California at Irvine, on June 8-10, 2021 and brought together 342 scientists, families, and industry representatives from over 25 countries to share the latest discoveries on underlying cellular and molecular mechanisms of T21, cognitive and behavioral changes, and comorbidities associated with Down syndrome, including Alzheimer's disease and Regression Disorder. Presentations of 91 cutting-edge abstracts reflecting neuroscience, neurology, model systems, psychology, biomarkers, and molecular and pharmacological therapeutic approaches demonstrate the compelling interest and continuing advancement toward innovating biomarkers and therapies aimed at ameliorating health conditions associated with T21.
Collapse
Affiliation(s)
- Eric D. Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lisi Flores-Aguilar
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, USA
| | - Benjamin Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Ann-Charlotte Granholm
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Stephanie Sherman
- Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| | - Victoria Puig
- Catalan Institute of Nanoscience and Nanotechnology, Barcelona, Spain
| | - Jonathan D. Santoro
- Neurological Institute, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - María Carmona-Iragui
- Hospital de la Santa Crue I Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, USA
| | - André Strydom
- Institute of Psychiatry, King's College London, London, UK
| | - Jorge Busciglio
- Neurobiology and Behavior School of Biological Sciences, University of California, Irvine, California, USA
| |
Collapse
|
41
|
Santoro J, Spinazzi N, Filipink R, Hayati-Rezvan P, Kammeyer R, Patel L, Sannar E, Dwyer L, Banerjee A, Khoshnood M, Jafarpour S, Boyd N, Partridge R, Gombolay G, Christy A, Real de Asua D, Del Carmen Ortega M, Manning M, Van Mater H, Worley G, Franklin C, Stanley M, Brown R, Capone G, Quinn E, Rafii M. Immunotherapy Responsiveness and Risk of Relapse in Down Syndrome Regression Disorder. RESEARCH SQUARE 2023:rs.3.rs-2521595. [PMID: 36824719 PMCID: PMC9949176 DOI: 10.21203/rs.3.rs-2521595/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Down syndrome regression disorder (DSRD) is a clinical symptom cluster consisting of neuropsychiatric regression without an identifiable cause. This study evaluated the clinical effectiveness of IVIg and evaluated clinical characteristics associated with relapse after therapy discontinuation. A prospective, multi-center, non-randomized, observational study was performed. Patients met criteria for DSRD and were treated with IVIg. All patients underwent a standardized wean off therapy after 9-12 months of treatment. Baseline, on therapy, and relapse scores of the Neuropsychiatric Inventory Total Score (NPITS), Clinical Global Impression-Severity (CGI-S), and the Bush-Francis Catatonia Rating Scale (BFCRS) were used to track clinical symptoms. Eighty-two individuals were enrolled in this study. Patients had lower BFCRS (MD: -6.68; 95% CI: -8.23, -5.14), CGI-S (MD: -1.27; 95% CI: -1.73, -0.81), and NPITS scores (MD: -6.50; 95% CI: -7.53, -5.47) while they were on therapy compared to baseline. Approximately 46% of the patients (n = 38) experienced neurologic relapse with wean of IVIg. Patients with neurologic relapse were more likely to have any abnormal neurodiagnostic study (χ2 = 11.82, p = 0.001), abnormal MRI (χ2 = 7.78, p = 0.005), and abnormal LP (χ2 = 5.45, p = 0.02), and a personal history of autoimmunity (OR: 6.11, p < 0.001) compared to patients without relapse. IVIg was highly effective in the treatment of DSRD. Individuals with a history of personal autoimmunity or neurodiagnostic abnormalities were more likely to relapse following weaning of immunotherapy, indicating the potential for, a chronic autoimmune etiology in some cases of DSRD.
Collapse
Affiliation(s)
| | | | | | | | - Ryan Kammeyer
- Kennedy-Krieger Institute and Johns Hopkins University
| | - Lina Patel
- Kennedy-Krieger Institute and Johns Hopkins University
| | - Elise Sannar
- Kennedy-Krieger Institute and Johns Hopkins University
| | - Luke Dwyer
- Kennedy-Krieger Institute and Johns Hopkins University
| | | | | | | | - Natalie Boyd
- Kennedy-Krieger Institute and Johns Hopkins University
| | | | | | | | | | | | | | | | - Gordon Worley
- Kennedy-Krieger Institute and Johns Hopkins University
| | | | - Maria Stanley
- Kennedy-Krieger Institute and Johns Hopkins University
| | - Ruth Brown
- Kennedy-Krieger Institute and Johns Hopkins University
| | - George Capone
- Kennedy-Krieger Institute and Johns Hopkins University
| | | | | |
Collapse
|
42
|
Viengkhou B, Hofer MJ. Breaking down the cellular responses to type I interferon neurotoxicity in the brain. Front Immunol 2023; 14:1110593. [PMID: 36817430 PMCID: PMC9936317 DOI: 10.3389/fimmu.2023.1110593] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Since their original discovery, type I interferons (IFN-Is) have been closely associated with antiviral immune responses. However, their biological functions go far beyond this role, with balanced IFN-I activity being critical to maintain cellular and tissue homeostasis. Recent findings have uncovered a darker side of IFN-Is whereby chronically elevated levels induce devastating neuroinflammatory and neurodegenerative pathologies. The underlying causes of these 'interferonopathies' are diverse and include monogenetic syndromes, autoimmune disorders, as well as chronic infections. The prominent involvement of the CNS in these disorders indicates a particular susceptibility of brain cells to IFN-I toxicity. Here we will discuss the current knowledge of how IFN-Is mediate neurotoxicity in the brain by analyzing the cell-type specific responses to IFN-Is in the CNS, and secondly, by exploring the spectrum of neurological disorders arising from increased IFN-Is. Understanding the nature of IFN-I neurotoxicity is a crucial and fundamental step towards development of new therapeutic strategies for interferonopathies.
Collapse
Affiliation(s)
- Barney Viengkhou
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
43
|
Cardiello JF, Westfall J, Dowell R, Allen MA. Characterizing Primary transcriptional responses to short term heat shock in paired fraternal lymphoblastoid lines with and without Down syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.524431. [PMID: 36712041 PMCID: PMC9882192 DOI: 10.1101/2023.01.17.524431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Heat shock stress induces genome wide changes in transcription regulation, activating a coordinated cellular response to enable survival. Using publicly available transcriptomic and proteomic data sets comparing individuals with and without trisomy 21, we noticed many heat shock genes are up-regulated in blood samples from individuals with trisomy 21. Yet no major heat shock response regulating transcription factor is encoded on chromosome 21, leaving it unclear why trisomy 21 itself would cause a heat shock response, or how it would impact the ability of blood cells to subsequently respond when faced with heat shock stress. To explore these issues in a context independent of any trisomy 21 associated co-morbidities or developmental differences, we characterized the response to heat shock of two lymphoblastoid cell lines derived from brothers with and without trisomy 21. To carefully compare the chromatin state and the transcription status of these cell lines, we measured nascent transcription, chromatin accessibility, and single cell transcript levels in the lymphoblastoid cell lines before and after acute heat shock treatment. The trisomy 21 cells displayed a more robust heat shock response after just one hour at 42°C than the matched disomic cells. We suggest multiple potential mechanisms for this increased heat shock response in lymphoblastoid cells with trisomy 21 including the possibility that cells with trisomy 21 may exist in a hyper-reactive state due to chronic stresses. Whatever the mechanism, abnormal heat shock response in individuals with Down syndrome may hobble immune responses during fever and contribute to health problems in these individuals.
Collapse
|
44
|
Sarver DC, Xu C, Velez LM, Aja S, Jaffe AE, Seldin MM, Reeves RH, Wong GW. Dysregulated systemic metabolism in a Down syndrome mouse model. Mol Metab 2023; 68:101666. [PMID: 36587842 PMCID: PMC9841171 DOI: 10.1016/j.molmet.2022.101666] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/14/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Trisomy 21 is one of the most complex genetic perturbations compatible with postnatal survival. Dosage imbalance arising from the triplication of genes on human chromosome 21 (Hsa21) affects multiple organ systems. Much of Down syndrome (DS) research, however, has focused on addressing how aneuploidy dysregulates CNS function leading to cognitive deficit. Although obesity, diabetes, and associated sequelae such as fatty liver and dyslipidemia are well documented in the DS population, only limited studies have been conducted to determine how gene dosage imbalance affects whole-body metabolism. Here, we conduct a comprehensive and systematic analysis of key metabolic parameters across different physiological states in the Ts65Dn trisomic mouse model of DS. METHODS Ts65Dn mice and euploid littermates were subjected to comprehensive metabolic phenotyping under basal (chow-fed) state and the pathophysiological state of obesity induced by a high-fat diet (HFD). RNA sequencing of liver, skeletal muscle, and two major fat depots were conducted to determine the impact of aneuploidy on tissue transcriptome. Pathway enrichments, gene-centrality, and key driver estimates were performed to provide insights into tissue autonomous and non-autonomous mechanisms contributing to the dysregulation of systemic metabolism. RESULTS Under the basal state, chow-fed Ts65Dn mice of both sexes had elevated locomotor activity and energy expenditure, reduced fasting serum cholesterol levels, and mild glucose intolerance. Sexually dimorphic deterioration in metabolic homeostasis became apparent when mice were challenged with a high-fat diet. While obese Ts65Dn mice of both sexes exhibited dyslipidemia, male mice also showed impaired systemic insulin sensitivity, reduced mitochondrial activity, and elevated fibrotic and inflammatory gene signatures in the liver and adipose tissue. Systems-level analysis highlighted conserved pathways and potential endocrine drivers of adipose-liver crosstalk that contribute to dysregulated glucose and lipid metabolism. CONCLUSIONS A combined alteration in the expression of trisomic and disomic genes in peripheral tissues contribute to metabolic dysregulations in Ts65Dn mice. These data lay the groundwork for understanding the impact of aneuploidy on in vivo metabolism.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cheng Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leandro M Velez
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew E Jaffe
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; The Lieber Institute for Brain Development, Baltimore, MD, USA; Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - Roger H Reeves
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
45
|
Araya P, Kinning KT, Coughlan C, Smith KP, Granrath RE, Enriquez-Estrada BA, Worek K, Sullivan KD, Rachubinski AL, Wolter-Warmerdam K, Hickey F, Galbraith MD, Potter H, Espinosa JM. IGF1 deficiency integrates stunted growth and neurodegeneration in Down syndrome. Cell Rep 2022; 41:111883. [PMID: 36577365 PMCID: PMC9876612 DOI: 10.1016/j.celrep.2022.111883] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 10/30/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022] Open
Abstract
Down syndrome (DS), the genetic condition caused by trisomy 21 (T21), is characterized by stunted growth, cognitive impairment, and increased risk of diverse neurological conditions. Although signs of lifelong neurodegeneration are well documented in DS, the mechanisms underlying this phenotype await elucidation. Here we report a multi-omics analysis of neurodegeneration and neuroinflammation biomarkers, plasma proteomics, and immune profiling in a diverse cohort of more than 400 research participants. We identified depletion of insulin growth factor 1 (IGF1), a master regulator of growth and brain development, as the top biosignature associated with neurodegeneration in DS. Individuals with T21 display chronic IGF1 deficiency downstream of growth hormone production, associated with a specific inflammatory profile involving elevated tumor necrosis factor alpha (TNF-α). Shorter children with DS show stronger IGF1 deficiency, elevated biomarkers of neurodegeneration, and increased prevalence of autism and other conditions. These results point to disruption of IGF1 signaling as a potential contributor to stunted growth and neurodegeneration in DS.
Collapse
Affiliation(s)
- Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kohl T Kinning
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christina Coughlan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Belinda A Enriquez-Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kayleigh Worek
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Section of Developmental Pediatrics, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kristine Wolter-Warmerdam
- Sie Center for Down Syndrome, Department of Pediatrics, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Francis Hickey
- Sie Center for Down Syndrome, Department of Pediatrics, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Huntington Potter
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
46
|
Krivega M, Stiefel CM, Storchova Z. Consequences of chromosome gain: A new view on trisomy syndromes. Am J Hum Genet 2022; 109:2126-2140. [PMID: 36459979 PMCID: PMC9808507 DOI: 10.1016/j.ajhg.2022.10.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Chromosome gains are detrimental for the development of the human embryo. As such, autosomal trisomies almost always result in spontaneous abortion, and the rare embryos surviving until live birth suffer from a plethora of pathological defects. There is no treatment currently available to ameliorate the consequences of trisomies, such as Down syndrome (trisomy of chromosome 21). Identifying the source of the phenotypes observed in cells with extra chromosomes is crucial for understanding the underlying molecular causes of trisomy syndromes. Although increased expression of the genes localized on the extra chromosome triggers several pathological phenotypes, an alternative model suggests that global, aneuploidy-associated changes in cellular physiology also contribute to the pathology. Here, we compare the molecular consequences of trisomy syndromes in vivo against engineered cell lines carrying various chromosome gains in vitro. We point out several phenotypes that are shared by variable trisomies and, therefore, might be caused by the presence of an extra chromosome per se, independent of its identity. This alternative view may provide useful insights for understanding Down syndrome pathology and open additional opportunities for diagnostics and treatments.
Collapse
Affiliation(s)
- Maria Krivega
- Reproduction Genetics, Department of Endocrinology and Infertility Disorders, Women Hospital, Heidelberg University, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany.
| | - Clara M Stiefel
- Department of Radiation Oncology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Zuzana Storchova
- Department of Molecular Genetics, Faculty of Biology, TU Kaiserslautern, Paul-Ehrlich-Str. 24, 67663 Kaiserslautern, Germany
| |
Collapse
|
47
|
Guild A, Fritch J, Patel S, Reinhardt A, Acquazzino M. Hemophagocytic lymphohistocytosis in trisomy 21: successful treatment with interferon inhibition. Pediatr Rheumatol Online J 2022; 20:104. [PMID: 36401314 PMCID: PMC9673190 DOI: 10.1186/s12969-022-00764-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/29/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening condition of immune dysregulation primarily driven by the cytokine interferon gamma. It can be either a genetic or acquired disorder associated with infection, malignancy, and rheumatologic disorders. Trisomy 21 can express a wide range of phenotypes which include immune dysregulation and shares inherent pathophysiology with a group of disorders termed interferonopathies. Knowledge of this overlap in seemingly unrelated conditions could provide a basis for future research, and most importantly, alternative therapeutic interventions in acute life threatening clinical scenarios. Herein, we describe two patients with trisomy 21 presenting with HLH that was refractory to conventional treatment. Both patients were successfully managed with novel interventions targeting the interferon pathway. CASE PRESENTATION We describe a 17-month-old male and 15-month-old female with trisomy 21 presenting with a myriad of signs and symptoms including fever, rash, cytopenias, and hyperferritinemia, both ultimately diagnosed with HLH. Each had relapsing, refractory HLH over time requiring several admissions to the hospital receiving conventional high dose corticosteroids and interleukin-1 inhibition therapy. Successful steroid-free remission was achieved after targeting interferon inhibition with emapalumab induction followed by long-term maintenance on baricitinib. CONCLUSION To our knowledge, these are the first reported cases of relapsed, refractory HLH in patients with trisomy 21 successfully treated with emapalumab and transitioned to a steroid-sparing regimen with oral baricitinib for maintenance therapy. Trisomy 21 autoimmunity and HLH are both thought to be driven by interferon gamma. Targeting therapy toward interferon signaling in both HLH and autoimmunity in trisomy 21 may have potential therapeutic benefits. Further investigation is needed to determine if trisomy 21 may predispose to the development of HLH given this common pathway.
Collapse
Affiliation(s)
- Allison Guild
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Jordan Fritch
- grid.266813.80000 0001 0666 4105Department of Pediatrics, Division of Hematology Oncology, University of Nebraska Medical Center, Omaha, NE USA
| | - Sachit Patel
- grid.266813.80000 0001 0666 4105Department of Pediatrics, Division of Hematology Oncology, University of Nebraska Medical Center, Omaha, NE USA
| | - Adam Reinhardt
- Department of Rheumatology, Boystown National Research Hospital, Omaha, NE USA
| | - Melissa Acquazzino
- grid.266813.80000 0001 0666 4105Department of Pediatrics, Division of Hematology Oncology, University of Nebraska Medical Center, Omaha, NE USA
| |
Collapse
|
48
|
Amin MA, Khan II, Nahin S, Bonna A, Afrin S, Hawlader MDH. COVID-19 hospitalization with later long COVID in a person with Down syndrome. Clin Case Rep 2022; 10:e6425. [PMID: 36245462 PMCID: PMC9540596 DOI: 10.1002/ccr3.6425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/27/2022] [Accepted: 09/20/2022] [Indexed: 11/12/2022] Open
Abstract
Viruses that induce pulmonary difficulties and auto-inflammation are more common in people with Down syndrome. They also have a higher number of comorbidities associated with a worse prognosis than the overall population. Adult patients with acute COVID-19 are increasingly being diagnosed with Long COVID. However, patients with Down syndrome with later long COVID-19 are the first example documented in Bangladesh.
Collapse
Affiliation(s)
- Mohammad Ashraful Amin
- Department of Public HealthNorth South UniversityDhakaBangladesh
- Public Health Professional Development Society (PPDS)DhakaBangladesh
| | | | - Sabrina Nahin
- Department of PhysiologyGreen Life Medical College HospitalDhakaBangladesh
| | - Atia Sharmin Bonna
- Public Health Professional Development Society (PPDS)DhakaBangladesh
- Public Health EpidemiologistHN & HIV Sector, Save the ChildrenDhakaBangladesh
| | - Sadia Afrin
- Department of Public HealthNorth South UniversityDhakaBangladesh
- Public Health Professional Development Society (PPDS)DhakaBangladesh
| | | |
Collapse
|
49
|
Treatment of Down Syndrome-Associated Arthritis with JAK Inhibition. Case Rep Rheumatol 2022; 2022:4889102. [PMID: 35879952 PMCID: PMC9308516 DOI: 10.1155/2022/4889102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022] Open
Abstract
Down syndrome (DS) results from a trisomy of chromosome 21, which causes immune dysregulation that leads to hyperactivation of interferon and Janus kinase (JAK) signaling. This results in complex medical abnormalities in the immune system and an increase in autoimmune and autoinflammatory conditions such as down syndrome-associated arthritis (DA). DA is an aggressive, destructive, inflammatory arthritis that is easily misdiagnosed and difficult to treat. Treatment commonly includes immunosuppressive therapy, but these are often associated with adverse effects and ineffectiveness. This case report outlines a 6-year-old male with DA that was successfully treated with the JAK inhibitor tofacitinib. Due to the aggressive nature of DA and poor response to many immunosuppressive therapies, this case report was created to increase awareness of JAK inhibition as an effective, well-tolerated treatment for DA.
Collapse
|
50
|
Jin M, Xu R, Wang L, Alam MM, Ma Z, Zhu S, Martini AC, Jadali A, Bernabucci M, Xie P, Kwan KY, Pang ZP, Head E, Liu Y, Hart RP, Jiang P. Type-I-interferon signaling drives microglial dysfunction and senescence in human iPSC models of Down syndrome and Alzheimer's disease. Cell Stem Cell 2022; 29:1135-1153.e8. [PMID: 35803230 DOI: 10.1016/j.stem.2022.06.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/27/2022] [Accepted: 06/09/2022] [Indexed: 12/17/2022]
Abstract
Microglia are critical in brain development and Alzheimer's disease (AD) etiology. Down syndrome (DS) is the most common genetic developmental disorder and risk factor for AD. Surprisingly, little information is available on the impact of trisomy of human chromosome 21 (Hsa21) on microglial functions during DS brain development and in AD in DS. Using induced pluripotent stem cell (iPSC)-based organoid and chimeric mouse models, we report that DS microglia exhibit an enhanced synaptic pruning function, which alters neuronal synaptic functions. In response to human brain tissue-derived pathological tau, DS microglia undergo cellular senescence and exhibit elevated type-I-interferon signaling. Mechanistically, knockdown of Hsa21-encoded type I interferon receptors, IFNARs, rescues the DS microglial phenotypes both during brain development and in response to pathological tau. Our findings provide in vivo evidence that human microglia respond to pathological tau by exhibiting dystrophic phenotypes. Targeting IFNARs may improve DS microglial functions and prevent senescence.
Collapse
Affiliation(s)
- Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Ranjie Xu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Le Wang
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Mahabub Maraj Alam
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Alessandra C Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Azadeh Jadali
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Matteo Bernabucci
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Kelvin Y Kwan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Ying Liu
- Department of Neurosurgery and Center for Stem Cell and Regenerative Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Center for Translational Science, Florida International University, Miami, FL 34987, USA
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|