1
|
Moretti NR, Silva ABND, Guimarães LV, Bezerra IDP, Taumaturgo JAC, Alves NGS, Kelly FA, Moraes FCAD. The prevalence of solid tumors and hematologic malignancies among patients with Down syndrome: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2025; 205:104558. [PMID: 39549894 DOI: 10.1016/j.critrevonc.2024.104558] [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: 08/19/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Patients with Down syndrome (DS) have a unique genetic and clinical profile that may increase the risk of cancer. METHODS A literature search on PubMed, Scopus, Web of Science, and the Cochrane databases was conducted, focusing on studies to investigate the prevalence of solid and hematologic tumors in DS. RESULTS Fifteen studies were included, encompassing 62,121 individuals with Down syndrome (DS). The overall prevalence of cancer in DS was 2.02 % (95 % CI: 1.63-2.50 %). The analysis of hematological tumors revealed a prevalence of 1.18 % (95 % CI: 0.86 % - 1.62 %) for leukemia, 0.86 % (95 % CI: 0.73 % - 1.01 %) for acute lymphoblastic leukemia, and 0.51 % (95 % CI: 0.29 % - 0.90 %) for acute myeloid leukemia. Among solid tumors, testicular cancer had the highest prevalence, at 0.22 % (95 % CI: 0.12 % - 0.43 %). CONCLUSIONS Our results highlight the need for targeted screening strategies, prevention strategies and treatment protocols among those with Down syndrome.
Collapse
Affiliation(s)
- Nayara Rozalem Moretti
- University of Western São Paulo, 700 José Bongiovani St Presidente Prudente, 19050-920, Brazil.
| | | | - Letícia Vieira Guimarães
- College of Medical and Health Sciences of Juiz de Fora, 200 Salvaterra Lane, Juiz de Fora 36033-003, Brazil
| | | | | | | | - Francinny Alves Kelly
- Dante Pazzanese Institute of Cardiology, 500 Dr. Dante Pazzanese Av., São Paulo 04012-909, Brazil
| | | |
Collapse
|
2
|
Xiang JM, Gao LL. Decisional conflict, anxiety, and social support among Chinese pregnant women making further prenatal testing decisions. J Reprod Infant Psychol 2025; 43:34-46. [PMID: 37403770 DOI: 10.1080/02646838.2023.2232380] [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: 01/09/2023] [Accepted: 06/26/2023] [Indexed: 07/06/2023]
Abstract
OBJECTIVE This study aimed to examine decisional conflict and identify its predictors in Chinese pregnant women who were making decisions about further prenatal testing after receiving a screening result of high-risk for Down syndrome. METHOD A cross-sectional study was conducted from September 2020 to July 2021 in Guangzhou, China. Two-hundred and sixty pregnant women receiving a screening result of high-risk for Down syndrome completed a questionnaire comprising the Decisional Conflict Scale, Self-rating Anxiety Scale, and Social Support Rating Scale. RESULTS The mean decisional conflict score was 28.8 ± 13.6, representing a moderate level. Advanced age (≥35 years), having a religious belief, not knowing about non-invasive or invasive prenatal testing, choosing NIPT for further prenatal testing, high levels of anxiety, and low levels of social support were significant predictors of decisional conflict, explaining 28.4% of its variance (F = 18.115, p < 0.001). CONCLUSIONS The results highlighted the necessity of assessing patients' decisional conflict and providing adequate interventions along the prenatal care trajectory. The results also showed that providing good support has an essential value for women by relieving their decisional conflict.
Collapse
Affiliation(s)
- Jia-Ming Xiang
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital,Central South University, Changsha, Hunan, China
| | - Ling-Ling Gao
- School of Nursing, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
3
|
Wedenoja S, Pihlajamäki M, Gissler M, Wedenoja J, Öhman H, Heinonen S, Kere J, Kääriäinen H, Tanner L. Infertility following trisomic pregnancies: A nationwide cohort study. Int J Gynaecol Obstet 2025; 168:326-332. [PMID: 39056516 PMCID: PMC11649879 DOI: 10.1002/ijgo.15828] [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: 06/06/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
OBJECTIVE To study whether gynecologic or reproductive disorders show association with trisomic conceptions. METHODS This nationwide cohort study utilized the Registry of Congenital Malformations to identify women who had a trisomic pregnancy (n = 5784), either with trisomy 13 (T13; n = 351), trisomy 18 (T18; n = 1065) or trisomy 21 (T21; n = 4369) from 1987 to 2018. We used the Finnish Maternity cohort to match the cases to population controls (n = 34 422) on the age, residence, and timing of pregnancy. These data were cross-linked to the ICD-10 diagnoses of the national Care Registry for Health Care data on specialized health care in Finland during 1996 to 2019. Both inflammatory (ICD-10 diagnoses: N70-N77) and noninflammatory disorders of the genital tract (N80-N98) were studied. Crude odds ratios (ORs) with 95% CIs were calculated for associations between diagnoses and trisomic conceptions. RESULTS The diagnosis of female infertility (N97) at any time was associated with trisomic conceptions (OR: 1.19, 95% CI: 1.08-1.32). In the subgroup analysis, this association was found for T18 (OR: 1.29, 95% CI: 1.03-1.61) and T21 (OR: 1.17, 95% CI: 1.04-1.32), but not for T13 (OR: 1.15, 95% CI: 0.75-1.72). When restricting the timing of the diagnosis of female infertility, an elevated OR was found only after the index pregnancy (OR: 1.81, 95% CI: 1.56-2.09). These increased odds for infertility after trisomic conceptions were observed both in women <35 years (T18 OR: 1.91, 95% CI: 1.21-3.00; T21 OR: 1.68, 95% CI: 1.31-2.14) and in women ≥35 years (T18 OR: 2.17, 95% CI: 1.40-3.33; T21 OR: 1.87; 95% CI: 1.47-2.39), but not after T13 conceptions. CONCLUSION Our observational data suggest a link between trisomic conceptions and subsequent diagnoses of infertility but do not demonstrate causality. These data implicate that partially similar mechanisms might predispose to trisomy and infertility, regardless of maternal age.
Collapse
Affiliation(s)
- Satu Wedenoja
- Information BrokersFinnish Institute for Health and WelfareHelsinkiFinland
- Obstetrics and GynecologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Stem Cells and Metabolism Research ProgramUniversity of HelsinkiHelsinkiFinland
- Folkhälsan Research CenterHelsinkiFinland
| | - Mika Pihlajamäki
- Information BrokersFinnish Institute for Health and WelfareHelsinkiFinland
| | - Mika Gissler
- Information BrokersFinnish Institute for Health and WelfareHelsinkiFinland
- Research Center for Child PsychiatryUniversity of TurkuTurkuFinland
- Region StockholmAcademic Primary Health Care CenterStockholmSweden
- Karolinska InstitutetDepartment of Molecular Medicine and SurgeryStockholmSweden
| | - Juho Wedenoja
- Department of OphthalmologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Hanna Öhman
- Biobank Borealis of Northern FinlandOulu University HospitalOuluFinland
- Faculty of MedicineUniversity of OuluOuluFinland
| | - Seppo Heinonen
- Obstetrics and GynecologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Juha Kere
- Stem Cells and Metabolism Research ProgramUniversity of HelsinkiHelsinkiFinland
- Folkhälsan Research CenterHelsinkiFinland
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Helena Kääriäinen
- Genomics and Biomarkers UnitNational Institute for Health and WelfareHelsinkiFinland
| | - Laura Tanner
- Department of Clinical GeneticsHelsinki University HospitalHelsinkiFinland
- Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland
| |
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
|
Piekoszewska-Ziętek P, Witt-Porczyk A, Turska-Szybka A, Olczak-Kowalczyk D. Hygienic behaviors and use of dental care in patients with genetic syndromes. Sci Rep 2024; 14:30756. [PMID: 39730420 DOI: 10.1038/s41598-024-80922-0] [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: 04/05/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
Patients with genetic syndromes require special dental attention because they have symptoms that promote plaque accumulation, dental erosion, dental caries and gingival diseases. The aim of the study was to assess hygienic behaviors, use of dental care and frequency of professional preventive procedures among Polish children and adolescents with Prader-Willi, Down, Angelman, Silver-Russell and Smith-Lemli-Opitz syndromes. Parents/legal guardians of children and adolescents with genetic syndromes were included. A questionnaire survey was conducted regarding socioeconomic factors, hygienic procedures performed at home and use of dental care as well as use of preventive treatments. The percentage of patients with genetic syndromes who received dental care was statistically significantly lower compared to the control group. Oral hygiene measures were most frequently used by participants with Silver-Russel syndrome, and less commonly by patients with Prader-Willi and Down syndrome. Dental treatment under general anesthesia was provided in 26 (38.2%) of the 68 children with genetic syndromes receiving dental care. Hygienic neglect and inadequate use of dental care due to limited access to certain preventive and therapeutic procedures among patients with genetic syndromes are worrying. It is necessary to educate and intensify caries prevention in this group of patients.
Collapse
Affiliation(s)
- Paula Piekoszewska-Ziętek
- Department of Paediatric Dentistry, Medical University of Warsaw, St. Binieckiego 6, 02-097, Warsaw, Poland.
| | | | - Anna Turska-Szybka
- Department of Paediatric Dentistry, Medical University of Warsaw, St. Binieckiego 6, 02-097, Warsaw, Poland
| | - Dorota Olczak-Kowalczyk
- Department of Paediatric Dentistry, Medical University of Warsaw, St. Binieckiego 6, 02-097, Warsaw, Poland.
| |
Collapse
|
6
|
Namikawa K, Pose-Méndez S, Köster RW. Genetic modeling of degenerative diseases and mechanisms of neuronal regeneration in the zebrafish cerebellum. Cell Mol Life Sci 2024; 82:26. [PMID: 39725709 DOI: 10.1007/s00018-024-05538-z] [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: 07/24/2024] [Revised: 10/11/2024] [Accepted: 12/01/2024] [Indexed: 12/28/2024]
Abstract
The cerebellum is a highly conserved brain compartment of vertebrates. Genetic diseases of the human cerebellum often lead to degeneration of the principal neuron, the Purkinje cell, resulting in locomotive deficits and socio-emotional impairments. Due to its relatively simple but highly conserved neuroanatomy and circuitry, these human diseases can be modeled well in vertebrates amenable for genetic manipulation. In the recent years, cerebellar research in zebrafish has contributed to understanding cerebellum development and function, since zebrafish larvae are not only molecularly tractable, but also accessible for high resolution in vivo imaging due to the transparency of the larvae and the ease of access to the zebrafish cerebellar cortex for microscopy approaches. Therefore, zebrafish is increasingly used for genetic modeling of human cerebellar neurodegenerative diseases and in particular of different types of Spinocerebellar Ataxias (SCAs). These models are well suited to address the underlying pathogenic mechanisms by means of in vivo cell biological studies. Furthermore, accompanying circuitry characterizations, physiological studies and behavioral analysis allow for unraveling molecular, structural and functional relationships. Moreover, unlike in mammals, zebrafish possess an astonishing ability to regenerate neuronal populations and their functional circuitry in the central nervous system including the cerebellum. Understanding the cellular and molecular processes of these regenerative processes could well serve to counteract acute and chronic loss of neurons in humans. Based on the high evolutionary conservation of the cerebellum these regeneration studies in zebrafish promise to open therapeutic avenues for counteracting cerebellar neuronal degeneration. The current review aims to provide an overview over currently existing genetic models of human cerebellar neurodegenerative diseases in zebrafish as well as neuroregeneration studies using the zebrafish cerebellum. Due to this solid foundation in cerebellar disease modeling and neuronal regeneration analysis, the zebrafish promises to become a popular model organism for both unraveling pathogenic mechanisms of human cerebellar diseases and providing entry points for therapeutic neuronal regeneration approaches.
Collapse
Affiliation(s)
- Kazuhiko Namikawa
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, 38106, Braunschweig, Germany
| | - Sol Pose-Méndez
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, 38106, Braunschweig, Germany
| | - Reinhard W Köster
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, 38106, Braunschweig, Germany.
| |
Collapse
|
7
|
Kanemaru E, Ichinose F. Essential role of sulfide oxidation in brain health and neurological disorders. Pharmacol Ther 2024; 266:108787. [PMID: 39719173 DOI: 10.1016/j.pharmthera.2024.108787] [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: 07/11/2024] [Revised: 11/21/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Hydrogen sulfide (H2S) is an environmental hazard well known for its neurotoxicity. In mammalian cells, H2S is predominantly generated by transsulfuration pathway enzymes. In addition, H2S produced by gut microbiome significantly contributes to the total sulfide burden in the body. Although low levels of H2S is believed to exert various physiological functions such as neurotransmission and vasomotor control, elevated levels of H2S inhibit the activity of cytochrome c oxidase (i.e., mitochondrial complex IV), thereby impairing oxidative phosphorylation. To protect the electron transport chain from respiratory poisoning by H2S, the compound is actively oxidized to form persulfides and polysulfides by a mitochondrial resident sulfide oxidation pathway. The reaction, catalyzed by sulfide:quinone oxidoreductase (SQOR), is the initial and critical step in sulfide oxidation. The persulfide species are subsequently oxidized to sulfite, thiosulfate, and sulfate by persulfide dioxygenase (ETHE1 or SDO), thiosulfate sulfurtransferase (TST), and sulfite oxidase (SUOX). While SQOR is abundantly expressed in the colon, liver, lung, and skeletal muscle, its expression is notably low in the brains of most mammals. Consequently, the brain's limited capacity to oxidize H2S renders it particularly sensitive to the deleterious effects of H2S accumulation. Impaired sulfide oxidation can lead to fatal encephalopathy, and the overproduction of H2S has been implicated in the developmental delays observed in Down syndrome. Our recent findings indicate that the brain's limited capacity to oxidize sulfide exacerbates its sensitivity to oxygen deprivation. The beneficial effects of sulfide oxidation are likely to be mediated not only by the detoxification of H2S but also by the formation of persulfide, which exerts cytoprotective effects through multiple mechanisms. Therefore, pharmacological agents designed to scavenge H2S and/or enhance persulfide levels may offer therapeutic potential against neurological disorders characterized by impaired or insufficient sulfide oxidation or excessive H2S production.
Collapse
Affiliation(s)
- Eiki Kanemaru
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
8
|
Zhou J, Zhong Y, Xu W. Effects of core stability exercises on balance ability of children and adolescents with intellectual disabilities: A systematic review and meta-analysis. PLoS One 2024; 19:e0314664. [PMID: 39700130 DOI: 10.1371/journal.pone.0314664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Children and adolescents with intellectual disabilities (IDs) are at risk of falls due to balance problems. One way to palliate balance deficits among this population is via core stability exercises. However, comprehensive studies that examine the effectiveness of core stability exercises in improving balance in this target population are lacking. OBJECTIVE This study aims to summarise and quantify the effectiveness of core stability exercises in improving the balance of this target population. METHODS This study followed PRISMA principles and conducted comprehensive searches in six academic databases (PubMed, Web of Science, Medline, Embase, Scopus, and the Cochrane Library) up to June 2023. The inclusion criteria were established via the PICOS framework. The risk of bias was assessed with the Cochrane risk-of-bias tool, and the certainty of the evidence was assessed via the GRADE approach. The meta-analysis was performed via RevMan 5.4, and for data that could not be pooled via meta-analysis, we used a narrative description of the results of each study. RESULTS Six studies of 1078 subjects were included. The findings revealed that core stability exercises improved the dynamic balance of children and adolescents with ID but had no significant effect on static (Hedges' g = 1.32, 95% CI [-0.41 to 3.06]) or static‒dynamic (Hedges' g = 1.35, 95% CI [-0.02 to 2.73]) balance compared with the control groups. The quality of evidence based on the GRADE approach was very low. CONCLUSIONS Core stability exercises may improve dynamic balance in children and adolescents with ID, but given the scarcity of studies included, definitive conclusions cannot yet be drawn. Although pooled analyses also highlighted improvements in static and static-dynamic balance with large effect sizes over active control groups, the results were not statistically significant and should be interpreted with caution given the wide confidence intervals. The heterogeneity among the identified studies and the limited number of eligible studies may reduce the reliability of the results, but these findings emphasise the need for additional research in this domain.
Collapse
Affiliation(s)
- Junjie Zhou
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang Province, China
| | - Yecheng Zhong
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang Province, China
| | - Wenhong Xu
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang Province, China
| |
Collapse
|
9
|
Sherman KM, Silveira CJ, Yan M, Yu L, Leon A, Klages K, White LG, Smith HM, Wolff SM, Falck A, Muneoka K, Brunauer R, Gaddy D, Suva LJ, Dawson LA. Male Down syndrome Ts65Dn mice have impaired bone regeneration. Bone 2024; 192:117374. [PMID: 39675408 DOI: 10.1016/j.bone.2024.117374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Trisomy of human chromosome 21 (Ts21) individuals present with a spectrum of low bone mineral density (BMD) that predisposes this vulnerable group to skeletal injuries. To determine the bone regenerative capacity of Down syndrome (DS) mice, male and female Dp16 and Ts65Dn DS mice underwent amputation of the digit tip (the terminal phalanx (P3)). This is a well-established mammalian model of bone regeneration that restores the amputated skeletal segment and all associated soft tissues. P3 amputation was performed in 8-week-old male and female DS mice and WT controls and followed by in vivo μCT, histology and immunofluorescence. Following P3 amputation, the bone degradation phase was attenuated in both Dp16 and Ts65Dn males. In Dp16 males, P3 regeneration was delayed but complete by 63 days post amputation (DPA); however, male Ts65Dn exhibited attenuated regeneration by 63 DPA. In both Dp16 and Ts65Dn female DS mice, P3 regenerates were indistinguishable from WT by 42 DPA. In Ts65Dn males, osteoclasts and eroded bone surface were significantly reduced, and osteoblast number significantly decreased in the regenerating digit. In Ts65Dn females, no significant differences were observed in any osteoclast or osteoblast parameter. Like Ts21 individuals and DS mice with sex differences in bone mass, these data expand the characteristic sexually dimorphism to include bone resorption and regeneration in response to skeletal injury in Ts65Dn mice. These observations suggest that sex differences contribute to the poor bone healing of DS and compound the increased risk of bone injury in the Ts21 population.
Collapse
Affiliation(s)
- Kirby M Sherman
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Catrina J Silveira
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Mingquan Yan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Ling Yu
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Abigail Leon
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Kasey Klages
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Lauren G White
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Hannah M Smith
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Sarah M Wolff
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Alyssa Falck
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Ken Muneoka
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Regina Brunauer
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America; LBG Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria.
| | - Dana Gaddy
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Larry J Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| | - Lindsay A Dawson
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, United States of America.
| |
Collapse
|
10
|
Bisogno S, Depciuch J, Gulzar H, Heber MF, Kobiałka M, Gąsior Ł, Bereta A, Pieczara A, Fic K, Musson R, Garcia Gamero G, Pardo Martinez M, Fornés Pérez A, Tatíčková M, Holubcova Z, Barańska M, Ptak GE. Female-age-dependent changes in the lipid fingerprint of the mammalian oocytes. Hum Reprod 2024; 39:2754-2767. [PMID: 39366679 PMCID: PMC11630086 DOI: 10.1093/humrep/deae225] [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: 01/15/2024] [Revised: 09/09/2024] [Indexed: 10/06/2024] Open
Abstract
STUDY QUESTION Can oocyte functionality be assessed by observing changes in their intracytoplasmic lipid droplets (LDs) profiles? SUMMARY ANSWER Lipid profile changes can reliably be detected in human oocytes; lipid changes are linked with maternal age and impaired developmental competence in a mouse model. WHAT IS KNOWN ALREADY In all cellular components, lipid damage is the earliest manifestation of oxidative stress (OS), which leads to a cascade of negative consequences for organelles and DNA. Lipid damage is marked by the accumulation of LDs. We hypothesized that impaired oocyte functionality resulting from aging and associated OS could be assessed by changes in LDs profile, hereafter called lipid fingerprint (LF). STUDY DESIGN, SIZE, DURATION To investigate if it is possible to detect differences in oocyte LF, we subjected human GV-stage oocytes to spectroscopic examinations. For this, a total of 48 oocytes derived from 26 young healthy women (under 33 years of age) with no history of infertility, enrolled in an oocyte donation program, were analyzed. Furthermore, 30 GV human oocytes from 12 women were analyzed by transmission electron microscopy (TEM). To evaluate the effect of oocytes' lipid profile changes on embryo development, a total of 52 C57BL/6 wild-type mice and 125 Gnpat+/- mice were also used. PARTICIPANTS/MATERIALS, SETTING, METHODS Human oocytes were assessed by label-free cell imaging via coherent anti-Stokes Raman spectroscopy (CARS). Further confirmation of LF changes was conducted using spontaneous Raman followed by Fourier transform infrared (FTIR) spectroscopies and TEM. Additionally, to evaluate whether LF changes are associated with developmental competence, mouse oocytes and blastocysts were evaluated using TEM and the lipid dyes BODIPY and Nile Red. Mouse embryonic exosomes were evaluated using flow cytometry, FTIR and FT-Raman spectroscopies. MAIN RESULTS AND THE ROLE OF CHANCE Here we demonstrated progressive changes in the LF of oocytes associated with the woman's age consisting of increased LDs size, area, and number. LF variations in oocytes were detectable also within individual donors. This finding makes LF assessment a promising tool to grade oocytes of the same patient, based on their quality. We next demonstrated age-associated changes in oocytes reflected by lipid peroxidation and composition changes; the accumulation of carotenoids; and alterations of structural properties of lipid bilayers. Finally, using a mouse model, we showed that LF changes in oocytes are negatively associated with the secretion of embryonic exosomes prior to implantation. Deficient exosome secretion disrupts communication between the embryo and the uterus and thus may explain recurrent implantation failures in advanced-age patients. LIMITATIONS, REASONS FOR CAUTION Due to differences in lipid content between different species' oocytes, the developmental impact of lipid oxidation and consequent LF changes may differ across mammalian oocytes. WIDER IMPLICATIONS OF THE FINDINGS Our findings open the possibility to develop an innovative tool for oocyte assessment and highlight likely functional connections between oocyte LDs and embryonic exosome secretion. By recognizing the role of oocyte LF in shaping the embryo's ability to implant, our original work points to future directions of research relevant to developmental biology and reproductive medicine. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by National Science Centre of Poland, Grants: 2021/41/B/NZ3/03507 and 2019/35/B/NZ4/03547 (to G.E.P.); 2022/44/C/NZ4/00076 (to M.F.H.) and 2019/35/N/NZ3/03213 (to Ł.G.). M.F.H. is a National Agency for Academic Exchange (NAWA) fellow (GA ULM/2019/1/00097/U/00001). K.F. is a Diamond Grant fellow (Ministry of Education and Science GA 0175/DIA/2019/28). The open-access publication of this article was funded by the Priority Research Area BioS under the program "Excellence Initiative - Research University" at the Jagiellonian University in Krakow. The authors declare no competing interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Simona Bisogno
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Joanna Depciuch
- Institute of Nuclear Physics, Polish Academy of Sciences, Kraków, Poland
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Hafsa Gulzar
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Kraków, Poland
| | - Maria Florencia Heber
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Michał Kobiałka
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Łukasz Gąsior
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Adrianna Bereta
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Anna Pieczara
- Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Kraków, Poland
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University in Krakow, Kraków, Poland
| | - Kinga Fic
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Richard Musson
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Kraków, Poland
| | - Gabriel Garcia Gamero
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Maria Pardo Martinez
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Alba Fornés Pérez
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Martina Tatíčková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zuzana Holubcova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Reprofit International, Brno, Czech Republic
| | - Małgorzata Barańska
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University in Krakow, Kraków, Poland
- Faculty of Chemistry, Jagiellonian University in Kraków, Kraków, Poland
| | - Grażyna Ewa Ptak
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| |
Collapse
|
11
|
Yehia Z, Silbereisen A, Koletsi D, Arabzadehtousi M, Tsilingaridis G, Bostanci N. Efficacy of periodontal treatment modalities in Down syndrome patients: a systematic review and meta-analysis. Evid Based Dent 2024; 25:213-214. [PMID: 39183214 PMCID: PMC11661967 DOI: 10.1038/s41432-024-01055-x] [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: 03/21/2024] [Accepted: 07/01/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVE The systematic review aimed to review the existing evidence, to identify and appraise the effectiveness of periodontal prevention and treatment modalities in individuals diagnosed with Down syndrome (DS) and to determine the estimates of the effects of implemented periodontal prevention and treatment strategies compared to chromosomally normal (CN) individuals. METHODOLOGY The systematic review was conducted and reported in conformity with the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) guidelines. The study protocol was registered in the Open Science Framework. Electronic and manual searches, in accordance with PICO framework and delineated inclusion/exclusion criteria, were conducted in multiple databases. RESULTS The initial search identified 11,704 studies. After removing duplicates, 9,048 remained. Title and abstract screening narrowed these to 281 for full-text review. Ultimately, 16 studies met the inclusion criteria, with 4 eligible for quantitative data synthesis. Results of the meta-analysis indicated that professional tooth cleaning in combination with oral hygiene reinforcement was less effective in the reduction of PPD in patients with DS compared to those without DS (Mean difference (MD): 0.23; 95% Confidence Interval (CI): 0.14 to 0.32; p < 0.001). DISCUSSION These findings suggest that conventional periodontal treatment is less effective in managing periodontitis in patients with DS. Thus, tailored periodontal care strategies that address the specific needs of individuals with DS should be implemented to improve treatment outcomes for this population The presence of moderate to high risk of bias in the included studies underscores the need for rigorously designed research that minimizes bias through effective blinding, randomization, control of confounding factors, and inclusion of diverse treatment outcomes to further investigate these associations. CONCLUSION Based on the best available evidence, professional tooth cleaning combined with oral hygiene instructions appears to be less effective in reducing pocket depths in individuals with DS compared to those without DS. https://doi.org/10.17605/OSF.IO/UXTCG.
Collapse
Affiliation(s)
- Zakaria Yehia
- Division of Periodontology and Oral Health, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Orthodontics, Folktandvården Stockholms län AB, Folktandvården Eastmaninstitutet, Stockholm, Sweden
| | - Angelika Silbereisen
- Division of Periodontology and Oral Health, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Despina Koletsi
- Clinic of Orthodontics and Pediatric Dentistry, Center of Dental Medicine, University of Zurich, Zurich, Switzerland
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA
| | - Mahla Arabzadehtousi
- Division of Periodontology and Oral Health, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Georgios Tsilingaridis
- Division of Orthodontics and Pediatric Dentistry, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre of Pediatric Oral Health, Stockholm, Sweden
| | - Nagihan Bostanci
- Division of Periodontology and Oral Health, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
12
|
Jhanji M, York EM, Lizarraga SB. The power of human stem cell-based systems in the study of neurodevelopmental disorders. Curr Opin Neurobiol 2024; 89:102916. [PMID: 39293245 DOI: 10.1016/j.conb.2024.102916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024]
Abstract
Neurodevelopmental disorders (NDDs) affect 15% of children and are usually associated with intellectual disability, seizures, and autistic behaviors, among other neurological presentations. Mutations in a wide spectrum of gene families alter key stages of human brain development, leading to defects in neural circuits or brain architecture. Studies in animal systems have provided important insights into the pathobiology of several NDDs. Human stem cell technologies provide a complementary system that allows functional manipulation of human brain cells during developmental stages that would otherwise be inaccessible during human fetal brain development. Therefore, stem cell-based models advance our understanding of human brain development by revealing human-specific mechanisms contributing to the broad pathogenesis of NDDs. We provide a comprehensive overview of the latest research on two and three-dimensional human stem cell-based models. First, we discuss convergent cellular and molecular phenotypes across different NDDs that have been revealed by human iPSC systems. Next, we examine the contribution of in vitro human neural systems to the development of promising therapeutic strategies. Finally, we explore the potential of stem cell systems to draw mechanistic insight for the study of sex dimorphism within NDDs.
Collapse
Affiliation(s)
- Megha Jhanji
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence RI 02906, USA; Center for Translational Neuroscience, Carney Brain Institute, Brown University, Providence RI 02906, USA
| | - Elisa M York
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence RI 02906, USA; Center for Translational Neuroscience, Carney Brain Institute, Brown University, Providence RI 02906, USA
| | - Sofia B Lizarraga
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence RI 02906, USA; Center for Translational Neuroscience, Carney Brain Institute, Brown University, Providence RI 02906, USA.
| |
Collapse
|
13
|
Harisinghani A, Cottrell C, Donelan K, Lam AD, Pulsifer M, Santoro SL. Practicalities (and real-life experiences) of dementia in adults with Down syndrome. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2024; 196:e32098. [PMID: 38967370 DOI: 10.1002/ajmg.c.32098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/06/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024]
Abstract
Adults with down syndrome (DS) have a lifetime dementia risk in excess of 95%, with a median age of onset of 55 years, due to trisomy 21. Co-occurring Alzheimer's disease (AD) has increased morbidity and mortality, and it is now recommended to screen for AD in all adults with DS beginning at 40 years of age. In this manuscript, we present two clinical cases of adults with DS who developed AD summarizing their medical histories, presenting symptoms, path to diagnosis and psychosocial aspects of care collected from retrospective chart review with caregiver consent. These two cases were chosen due to their complexity and interwoven nature of the medical and psychosocial aspects, and highlight the complexity and nuance of caring for patients with DS and AD.
Collapse
Affiliation(s)
- Ayesha Harisinghani
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Clorinda Cottrell
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Karen Donelan
- The Mongan Institute, Survey Research Unit, MGH, Boston, Massachusetts, USA
- Heller School for Social Policy and Management, Brandeis University, Waltham, Massachusetts, USA
| | - Alice D Lam
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Margaret Pulsifer
- Psychology Assessment Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephanie L Santoro
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
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
|
15
|
Huang T, Fakurazi S, Cheah PS, Ling KH. Chromosomal and cellular therapeutic approaches for Down syndrome: A research update. Biochem Biophys Res Commun 2024; 735:150664. [PMID: 39260337 DOI: 10.1016/j.bbrc.2024.150664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
In individuals with Down syndrome (DS), an additional HSA21 chromosome copy leads to the overexpression of a myriad of HSA21 genes, disrupting the transcription of the entire genome. This dysregulation in transcription and post-transcriptional modifications contributes to abnormal phenotypes across nearly all tissues and organs in DS individuals. The array of severe clinical symptoms associated with trisomy 21 poses a considerable challenge in the quest for a cure for DS. Fortunately, a wealth of research suggests that chromosome therapy, hinging on cutting-edge genome editing technologies, can potentially eliminate the extra copy of the human chromosome 21. Genome editing tools have demonstrated their efficacy in restoring trisomy to a normal diploid state in vitro DS cell models. Furthermore, we delve into the noteworthy findings in cellular therapy for DS, with recent studies showcasing the increasing feasibility of strategies involving stem cells and CAR T-cells to address corresponding clinical phenotypes.
Collapse
Affiliation(s)
- Tan Huang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Malaysian Research Institute on Ageing (MyAgeing(TM)), Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Malaysian Research Institute on Ageing (MyAgeing(TM)), Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
16
|
Lanzillotta S, Esteve D, Lanzillotta C, Tramutola A, Lloret A, Forte E, Pesce V, Picca A, Di Domenico F, Perluigi M, Barone E. Altered Mitochondrial Unfolded Protein Response and Protein Quality Control promote oxidative distress in Down Syndrome brain. Free Radic Biol Med 2024:S0891-5849(24)01077-3. [PMID: 39586382 DOI: 10.1016/j.freeradbiomed.2024.11.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
Down Syndrome (DS) is a genetic disorder caused by the presence of an extra copy of chromosome 21, and leading to various developmental and cognitive defects. A critical feature of DS is the occurrence of oxidative distress particularly in the brain, which exacerbates neurodevelopmental processes. Mitochondria play a crucial role in cell energy metabolism and their impairment is one of the major causes of oxidative distress in several pathologies. Hence, this study investigates mitochondrial proteostasis by the mean of the mitochondrial Unfolded Protein Response (UPRmt) and the mitochondrial protein quality control (MQC) mechanisms in the context of DS, focusing on their implications in redox homeostasis in brain development. We analyzed key UPRmt markers and mitochondrial function in the frontal cortex isolated fromTs2Cje mice, a model for DS, across different developmental stages. Our results demonstrate significant alterations in UPRmt markers, particularly at postnatal day 0 (P0) and 1 month (1M). These changes indicate early UPRmt activation, primarily driven by the ATF5/GRP75 axis, although compromised by reduced levels of other components. Impaired UPRmt correlates with decreased mitochondrial activity, evidenced by reduced oxygen consumption rates and altered expression of OXPHOS complexes. Additionally, elevated oxidative stress markers such as 3-nitrotyrosine (3-NT), 4-hydroxynonenal (HNE), and protein carbonyls (PC) were observed, linking mitochondrial dysfunction to increased oxidative damage. Defects of MQC, including disrupted biogenesis, increased fission, and the activation of mitophagy were evident mostly at P0 and 1M consistent with UPRmt activation. Principal Component Analysis revealed distinct phenotypic differences between Ts2Cje and control mice, driven by these molecular alterations. Our findings underscore the critical role of UPRmt and MQC in DS brain development, highlighting potential therapeutic targets to mitigate mitochondrial dysfunction and oxidative distress, thereby alleviating some of the neurodevelopmental and cognitive impairments associated with DS.
Collapse
Affiliation(s)
- Simona Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Daniel Esteve
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain; Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Ana Lloret
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Elena Forte
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Vito Pesce
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
17
|
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
|
18
|
Lee SE, Baxter LL, Duran MI, Morris SD, Mosley IA, Fuentes KA, Pennings JLA, Guedj F, Bianchi DW. Analysis of genotype effects and inter-individual variability in iPSC-derived trisomy 21 neural progenitor cells. Hum Mol Genet 2024:ddae160. [PMID: 39533854 DOI: 10.1093/hmg/ddae160] [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: 07/29/2024] [Revised: 10/09/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Trisomy of human chromosome 21 (T21) gives rise to Down syndrome (DS), the most frequent live-born autosomal aneuploidy. T21 triggers genome-wide transcriptomic alterations that result in multiple atypical phenotypes with highly variable penetrance and expressivity in individuals with DS. Many of these phenotypes, including atypical neurodevelopment, emerge prenatally. To enable in vitro analyses of the cellular and molecular mechanisms leading to the neurological alterations associated with T21, we created and characterized a panel of genomically diverse T21 and euploid induced pluripotent stem cells (iPSCs). We subsequently differentiated these iPSCs to generate a panel of neural progenitor cells (NPCs). Alongside characterizing genotype effects from T21, we found that T21 NPCs showed inter-individual variability in growth rates, oxidative stress, senescence characteristics, and gene and protein expression. Pathway enrichment analyses of T21 NPCs identified vesicular transport, DNA repair, and cellular response to stress pathways. These results demonstrate T21-associated variability at the cellular level and suggest that cell lines from individuals with DS should not solely be analyzed as a homogenous population. Examining large cohorts of genetically diverse samples may more fully reveal the effects of aneuploidy on transcriptomic and phenotypic characteristics in T21 cell types. A panel of genomically diverse T21 and euploid induced pluripotent stem cells (iPSCs) were created and subsequently differentiated into neural progenitor cells (NPCs). T21 NPCs showed reduced growth, increased oxidative stress, and inter-individual variability in gene and protein expression. This inter-individual variability suggests that studies with large cohorts of genetically diverse T21 samples may more fully reveal the effects of aneuploidy.
Collapse
Affiliation(s)
- Sarah E Lee
- Prenatal Genomics and Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, 35A Convent Drive Bethesda, MD 20892, United States
| | - Laura L Baxter
- Prenatal Genomics and Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, 35A Convent Drive Bethesda, MD 20892, United States
| | - Monica I Duran
- Prenatal Genomics and Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, 35A Convent Drive Bethesda, MD 20892, United States
| | - Samuel D Morris
- Prenatal Genomics and Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, 35A Convent Drive Bethesda, MD 20892, United States
| | - Iman A Mosley
- Prenatal Genomics and Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, 35A Convent Drive Bethesda, MD 20892, United States
| | - Kevin A Fuentes
- Prenatal Genomics and Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, 35A Convent Drive Bethesda, MD 20892, United States
| | - Jeroen L A Pennings
- Center for Health Protection, National Institute for Public Health and the Environment, P.O. Box 1, Bilthoven, BA 3720, the Netherlands
| | - Faycal Guedj
- Prenatal Genomics and Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, 35A Convent Drive Bethesda, MD 20892, United States
| | - Diana W Bianchi
- Prenatal Genomics and Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, 35A Convent Drive Bethesda, MD 20892, United States
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 31 Center Drive, Bethesda, MD 20892, United States
| |
Collapse
|
19
|
Huang T, Lam XJ, Lim CT, Jusoh N, Fakurazi S, Cheah PS, Ling KH. Understanding perspectives and research trends in Down syndrome neuropathogenesis: A bibliometric analysis. JOURNAL OF INTELLECTUAL DISABILITIES : JOID 2024:17446295241299160. [PMID: 39533897 DOI: 10.1177/17446295241299160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Down syndrome (DS), characterised by compromised brain development and intellectual challenges, often manifests Alzheimer's disease (AD) -like symptoms. Utilising the Web of Science Core Collection (WOSCC) database from January 1, 2000, to July 31, 2023, we conducted a comprehensive bibliometric analysis using VOSviewer, CiteSpace, and the R package "bibliometrix." Analyses included co-authorship, co-citation, co-occurrence, cooperative network, reference, and keyword burst citation. Analysing 5,082 papers, the U.S. demonstrated prominence with the highest number of research organisations and citations. Keyword analysis revealed promising research areas, including "Alzheimer's disease," "development," "inflammation," and "neurogenesis". This 22-year survey of the brain with trisomy 21 research unveils key trends, contributors, and focal areas in DS neuropathogenesis. Notably, Alzheimer 's-related genes and proteins play a pervasive role in DS neuropathological processes across patients' lifespans. The study contributes foundational knowledge for advancing research and care in the DS neuropathogenesis domain.
Collapse
Affiliation(s)
- Tan Huang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Xin-Jieh Lam
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Chong-Teik Lim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Norhazlin Jusoh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
- Malaysian Research Institute on Ageing (MyAgeing®), Universiti Putra Malaysia, Malaysia
| |
Collapse
|
20
|
Castellani RJ, Jamshidi P, Plascencia-Villa G, Perry G. The Amyloid Cascade Hypothesis: A Conclusion in Search of Support. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00407-3. [PMID: 39532243 DOI: 10.1016/j.ajpath.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
The amyloid cascade hypothesis as the etiological underpinning of Alzheimer disease (AD) is supported by a large body of literature, the most influential of which are genetic studies of the 1980s and 1990s. Other evidence includes the neuropathology of Down syndrome, apparent toxicity of oligomeric amyloid-β (Aβ), interactions with apolipoprotein E, and the analogy of cardiac amyloidosis. On the other hand, there is considerable phenotypic heterogeneity among the rare familial AD kindreds, which tempers extrapolation to sporadic AD. Oligomer biology is still in the theoretical realm, with no clinical validation. Apolipoprotein E support for the amyloid cascade and other inferences from the literature are somewhat circular in their logic. Analogy with amyloidoses might also consider secondary amyloidosis, driven by systemic inflammation and treated by treating the underlying etiology. Much of the remaining literature supporting the amyloid cascade is dominated by hypothesis-generating studies. Importantly, we now have a developing evidence base from controlled clinical trials that can potentially inform the issue of Aβ as a cause or driver of disease in sporadic AD. Emerging data provide clear evidence of target engagement. Clinical outcome, however, has been either marginally positive or similar to placebo. Assuming these findings hold, it appears that Aβ neither drives nor mitigates the disease process.
Collapse
Affiliation(s)
- Rudy J Castellani
- Division of Neuropathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Pouya Jamshidi
- Division of Neuropathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Germán Plascencia-Villa
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas
| |
Collapse
|
21
|
Maure-Blesa L, Rodríguez-Baz I, Carmona-Iragui M, Fortea J. What Can We Learn About Alzheimer's Disease from People with Down Syndrome? Curr Top Behav Neurosci 2024. [PMID: 39509049 DOI: 10.1007/7854_2024_546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Down syndrome (DS) is the most frequent cause of intellectual disability of genetic origin, estimated to affect about 1 in 700 babies born worldwide (CDC 2023). In Europe and the United States, current estimates indicate a population prevalence of 5.6 and 6.7 per 10,000 individuals, respectively, which translates to more than 200,000 people in the United States, more than 400,000 people in Europe, and approximately six million worldwide. Advances in healthcare and the treatment of accompanying conditions have significantly prolonged the lifespan of those with DS over the past 50 years. Consequently, there is a pressing need to address the challenges associated with ageing among this population, with Alzheimer's disease (AD) being the primary concern. In this chapter, we will review the significance of studying this population to understand AD biology, the insights gained on AD in DS (DSAD), and how this knowledge can help us understand the AD not only in DS but also in the general population. We will conclude by exploring the objectives that remain to be accomplished.
Collapse
Affiliation(s)
- Lucia Maure-Blesa
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Iñigo Rodríguez-Baz
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain.
| |
Collapse
|
22
|
Mouli K, Liopo AV, Suva LJ, Olson KR, McHugh EA, Tour JM, Derry PJ, Kent TA. SOD1 Is an Integral Yet Insufficient Oxidizer of Hydrogen Sulfide in Trisomy 21 B Lymphocytes and Can Be Augmented by a Pleiotropic Carbon Nanozyme. Antioxidants (Basel) 2024; 13:1361. [PMID: 39594503 PMCID: PMC11591310 DOI: 10.3390/antiox13111361] [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: 10/10/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Down syndrome (DS) is a multisystemic disorder that includes accelerated aging caused by trisomy 21. In particular, overexpression of cystathionine-β-synthase (CBS) is linked to excess intracellular hydrogen sulfide (H2S), a mitochondrial toxin at higher concentrations, which impairs cellular viability. Concurrent overexpression of superoxide dismutase 1 (SOD1) may increase oxidative stress by generating excess hydrogen peroxide (H2O2) while also mitigating the toxic H2S burden via a non-canonical sulfide-oxidizing mechanism. We investigated the phenotypic variability in basal H2S levels in relation to DS B lymphocyte cell health and SOD1 in H2S detoxification. The H2S levels were negatively correlated with the DS B lymphocyte growth rates but not with CBS protein. Pharmacological inhibition of SOD1 using LCS-1 significantly increased the H2S levels to a greater extent in DS cells while also decreasing the polysulfide products of H2S oxidation. However, DS cells exhibited elevated H2O2 and lipid peroxidation, representing potential toxic consequences of SOD1 overexpression. Treatment of DS cells with a pleiotropic carbon nanozyme (pleozymes) decreased the total oxidative stress and reduced the levels of the H2S-generating enzymes CBS and 3-mercaptopyruvate sulfurtransferase (MPST). Our results indicate that pleozymes may bridge the protective and deleterious effects of DS SOD1 overexpression on H2S metabolism and oxidative stress, respectively, with cytoprotective benefits.
Collapse
Affiliation(s)
- Karthik Mouli
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (K.M.); (A.V.L.)
| | - Anton V. Liopo
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (K.M.); (A.V.L.)
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
| | - Larry J. Suva
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA;
| | - Kenneth R. Olson
- Department of Physiology, Indiana University School of Medicine South Bend, South Bend, IN 46617, USA;
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Emily A. McHugh
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
| | - James M. Tour
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
- Department of Materials Science and NanoEngineering, Rice University, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, Houston, TX 77005, USA
- NanoCarbon Center and the Rice Advanced Materials Institute, Rice University, Houston, TX 77005, USA
| | - Paul J. Derry
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (K.M.); (A.V.L.)
- School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Thomas A. Kent
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (K.M.); (A.V.L.)
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
| |
Collapse
|
23
|
Zhong G, Chang X, Xie W, Zhou X. Targeted protein degradation: advances in drug discovery and clinical practice. Signal Transduct Target Ther 2024; 9:308. [PMID: 39500878 PMCID: PMC11539257 DOI: 10.1038/s41392-024-02004-x] [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: 05/08/2024] [Revised: 08/19/2024] [Accepted: 09/28/2024] [Indexed: 11/08/2024] Open
Abstract
Targeted protein degradation (TPD) represents a revolutionary therapeutic strategy in disease management, providing a stark contrast to traditional therapeutic approaches like small molecule inhibitors that primarily focus on inhibiting protein function. This advanced technology capitalizes on the cell's intrinsic proteolytic systems, including the proteasome and lysosomal pathways, to selectively eliminate disease-causing proteins. TPD not only enhances the efficacy of treatments but also expands the scope of protein degradation applications. Despite its considerable potential, TPD faces challenges related to the properties of the drugs and their rational design. This review thoroughly explores the mechanisms and clinical advancements of TPD, from its initial conceptualization to practical implementation, with a particular focus on proteolysis-targeting chimeras and molecular glues. In addition, the review delves into emerging technologies and methodologies aimed at addressing these challenges and enhancing therapeutic efficacy. We also discuss the significant clinical trials and highlight the promising therapeutic outcomes associated with TPD drugs, illustrating their potential to transform the treatment landscape. Furthermore, the review considers the benefits of combining TPD with other therapies to enhance overall treatment effectiveness and overcome drug resistance. The future directions of TPD applications are also explored, presenting an optimistic perspective on further innovations. By offering a comprehensive overview of the current innovations and the challenges faced, this review assesses the transformative potential of TPD in revolutionizing drug development and disease management, setting the stage for a new era in medical therapy.
Collapse
Affiliation(s)
- Guangcai Zhong
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xiaoyu Chang
- School of Pharmaceutical Sciences, Pingyuan Laboratory, Zhengzhou University, Zhengzhou, 450001, China
| | - Weilin Xie
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
24
|
Russell JK, Conley AC, Wilson JE, Newhouse PA. Cholinergic System Structure and Function Changes in Individuals with Down Syndrome During the Development of Alzheimer's Disease. Curr Top Behav Neurosci 2024. [PMID: 39485646 DOI: 10.1007/7854_2024_523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Adults with Down syndrome represent the population with the highest risk of developing Alzheimer's disease worldwide. The cholinergic system is known to decline in Alzheimer's disease, with this decline responsible for many of the cognitive deficits that develop. The integrity of the cholinergic system across the lifespan in individuals with Down syndrome is not well characterized. Small fetal and infant post-mortem studies suggest an intact cholinergic projection system with a potential reduction in cholinergic receptors, while post-mortem studies in adults with Down syndrome reveal an age-related decrease in cholinergic integrity. Advances in magnetic resonance imaging (MRI) and positron emission tomography (PET) over the last 20 years have allowed for studies investigating the changes in cholinergic integrity across aging and during the development of Alzheimer's disease. One large cross-sectional study demonstrated reduced cholinergic basal forebrain volume measured by MRI associated with increasing Alzheimer's disease pathology. In a small cohort of adults with Down syndrome, we have recently reported that PET measures of cholinergic integrity negatively correlated with amyloid accumulation. New disease-modifying treatments for Alzheimer's disease and treatments under development for Alzheimer's disease in Down syndrome have the potential to preserve the cholinergic system, while treatments targeting the cholinergic system directly may be used in conjunction with disease-modifying therapies to improve cognitive function further. A greater understanding of cholinergic neuronal and receptor integrity across the lifespan in individuals with Down syndrome will provide insights as to when targeting the cholinergic system is an appropriate therapeutic option and, in the future, maybe a valuable screening tool to identify individuals that would most benefit from cholinergic interventions.
Collapse
Affiliation(s)
- Jason K Russell
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander C Conley
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jo Ellen Wilson
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Tennessee Valley Health System, Nashville, TN, USA
| | - Paul A Newhouse
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Tennessee Valley Health System, Nashville, TN, USA.
| |
Collapse
|
25
|
Martins KR, Alves FA, Silva LRD, Silva LOAD, Segundo GRS. Different immunological patterns of Down syndrome patients with and without recurrent infections. J Pediatr (Rio J) 2024; 100:653-659. [PMID: 39053889 PMCID: PMC11662752 DOI: 10.1016/j.jped.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE Individuals with Down Syndrome (DS) exhibit a higher susceptibility to infections, suggesting potential immunological alterations within this population. Consequently, this study aims to assess the immune response profile in children with DS to identify potential immune dysfunctions associated with recurrent infections. METHODS The authors conducted a retrospective analysis involving 49 DS patients, examining various epidemiological, clinical, cytogenetic, and laboratory variables. The study's sample comprised patients aged 2-20 years, with a predominance of males. These patients were categorized into two groups based on the presence or absence of recurrent infections, as indicated by the Jeffrey Modell Foundation alert signs. RESULTS Immunoglobulin (Ig) A, G, and M levels were deemed normal, although individuals with DS experiencing recurrent infections exhibited significantly lower IgA levels. Additionally, CD3, CD4, CD8, and CD19 lymphocyte counts were found to be within normal ranges, with no significant differences between the two groups. While overall data indicated normal seroconversion levels of pneumococcal polysaccharide antibodies, a notable impairment in seroconversion was observed among DS patients with recurrent infections compared to those without such infections. CONCLUSION The deficiency of anti-polysaccharide antibodies in individuals with DS may constitute an important immunological comorbidity. Therefore, it warrants further investigation, particularly among individuals with recurrent infections.
Collapse
Affiliation(s)
- Kamila Rosa Martins
- Universidade Federal de Uberlândia, Hospital de Clínicas, Uberlândia, MG, Brazil
| | | | - Luiz Roberto da Silva
- Universidade Federal de Uberlândia, Faculdade de Medicina, Departamento de Pediatria, Uberlândia, MG, Brazil
| | | | | |
Collapse
|
26
|
Fucà E, Costanzo F, Vicari S. Characterising repetitive behaviours in children and adolescents with Down syndrome. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2024; 68:1287-1300. [PMID: 39105267 DOI: 10.1111/jir.13179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Individuals with intellectual disability, including people with Down syndrome (DS), often exhibit restricted and repetitive behaviours (RRBs). However, RRBs have not been deeply characterised in children and adolescents with DS. METHOD The study encompassed a cohort of 151 participants aged 4 to 18 years with DS. RRBs were assessed utilising the Repetitive Behaviour Scale-Revised. Additionally, data pertaining to cognitive and adaptive functioning, linguistic abilities, sleep patterns and emotional/behavioural issues were gathered. RESULTS Self-injurious behaviours were reported less frequently whereas parents most commonly endorsed items related to behaviours associated with the need for sameness and ritualistic behaviours. We observed very few gender differences, whereas some age-related differences emerged, with adolescents exhibiting higher scores in items related with higher-level RRBs. The analysis of the association between RRBs and clinical features revealed that RRBs were associated with parent-reported sleep difficulties, as well as with internalising and externalising problems. We also observed a negative correlation with IQ whereas associations with adaptive skills emerged mainly for lower-level RRBs, such as motor stereotypies. Finally, RRBs were negatively associated with linguistic abilities, both expressive and receptive. CONCLUSIONS RRBs in children and adolescents with DS are of significant clinical interest due to their associations with various clinical dimensions. Therefore, psychological and neuropsychiatric assessment should include an accurate evaluation of RRBs for young people with DS.
Collapse
Affiliation(s)
- E Fucà
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - F Costanzo
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - S Vicari
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Life Science and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
27
|
Anshu K, Kristensen K, Godar SP, Zhou X, Hartley SL, Litovsky RY. Speech Recognition and Spatial Hearing in Young Adults With Down Syndrome: Relationships With Hearing Thresholds and Auditory Working Memory. Ear Hear 2024; 45:1568-1584. [PMID: 39090791 PMCID: PMC11493531 DOI: 10.1097/aud.0000000000001549] [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] [Indexed: 08/04/2024]
Abstract
OBJECTIVES Individuals with Down syndrome (DS) have a higher incidence of hearing loss (HL) compared with their peers without developmental disabilities. Little is known about the associations between HL and functional hearing for individuals with DS. This study investigated two aspects of auditory functions, "what" (understanding the content of sound) and "where" (localizing the source of sound), in young adults with DS. Speech reception thresholds in quiet and in the presence of interferers provided insight into speech recognition, that is, the "what" aspect of auditory maturation. Insights into "where" aspect of auditory maturation were gained from evaluating speech reception thresholds in colocated versus separated conditions (quantifying spatial release from masking) as well as right versus left discrimination and sound location identification. Auditory functions in the "where" domain develop during earlier stages of cognitive development in contrast with the later developing "what" functions. We hypothesized that young adults with DS would exhibit stronger "where" than "what" auditory functioning, albeit with the potential impact of HL. Considering the importance of auditory working memory and receptive vocabulary for speech recognition, we hypothesized that better speech recognition in young adults with DS, in quiet and with speech interferers, would be associated with better auditory working memory ability and receptive vocabulary. DESIGN Nineteen young adults with DS (aged 19 to 24 years) participated in the study and completed assessments on pure-tone audiometry, right versus left discrimination, sound location identification, and speech recognition in quiet and with speech interferers that were colocated or spatially separated. Results were compared with published data from children and adults without DS and HL, tested using similar protocols and stimuli. Digit Span tests assessed auditory working memory. Receptive vocabulary was examined using the Peabody Picture Vocabulary Test Fifth Edition. RESULTS Seven participants (37%) had HL in at least 1 ear; 4 individuals had mild HL, and 3 had moderate HL or worse. Participants with mild or no HL had ≥75% correct at 5° separation on the discrimination task and sound localization root mean square errors (mean ± SD: 8.73° ± 2.63°) within the range of adults in the comparison group. Speech reception thresholds in young adults with DS were higher than all comparison groups. However, spatial release from masking did not differ between young adults with DS and comparison groups. Better (lower) speech reception thresholds were associated with better hearing and better auditory working memory ability. Receptive vocabulary did not predict speech recognition. CONCLUSIONS In the absence of HL, young adults with DS exhibited higher accuracy during spatial hearing tasks as compared with speech recognition tasks. Thus, auditory processes associated with the "where" pathways appear to be a relative strength than those associated with "what" pathways in young adults with DS. Further, both HL and auditory working memory impairments contributed to difficulties in speech recognition in the presence of speech interferers. Future larger-sized samples are needed to replicate and extend our findings.
Collapse
Affiliation(s)
- Kumari Anshu
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Kayla Kristensen
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Shelly P. Godar
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Xin Zhou
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
- Currently at The Chinese University of Hong Kong, Hong Kong
| | - Sigan L. Hartley
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
- School of Human Ecology, University of Wisconsin–Madison, Madison, WI, USA
| | - Ruth Y. Litovsky
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
- Department of Communication Sciences and Disorders, University of Wisconsin–Madison, Madison, WI, USA
| |
Collapse
|
28
|
Zhu B, Yin B, Li H, Chu X, Mi Z, Sun Y, Yuan X, Chen R, Ma Z. A prediction model for gestational diabetes mellitus based on steroid hormonal changes in early and mid-down syndrome screening: A multicenter longitudinal study. Diabetes Res Clin Pract 2024; 217:111865. [PMID: 39307357 DOI: 10.1016/j.diabres.2024.111865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/31/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Steroid hormones (SH) during pregnancy are associated with the development of gestational diabetes mellitus (GDM). Early and mid-Down syndrome screening is used to assess the risk of Down syndrome in the fetus. It is unclear whether changes in SH during this period can be used as an early predictor of GDM. METHODS This study was a multicenter, longitudinal cohort study. GDM is diagnosed by an oral glucose tolerance test (OGTT) between 24 and 28 weeks of gestation. We measured SH levels at early and mid-Down syndrome screening, respectively. Based on the SH changes, logistic regression analysis was used to construct a prediction model for GDM. Finally, evaluated the model's predictive performance by creating a receiver operating characteristic curve (ROC) and performing external validation. RESULTS This study enrolled 193 pregnant women (discovery cohort, n = 157; validation cohort, n = 36). SH changes occur dynamically after pregnancy. At early Down syndrome screening, only cortisol (F) (p < 0.05, 95 % CI 4780.95-46083.68) was elevated in GDM. At mid-Down syndrome screening, free testosterone (FT) (p < 0.01, 95 % CI 0.10-0.55) and estradiol (E2) (p < 0.05, 95 % CI 203.55-1784.78) were also significantly elevated. There were significant differences in the rates of change in E2 (Fold change (FC) = 1.3425, p = 0.0072), albumin (ALB) (FC=1.5759, p = 0.0117), and dihydrotestosterone (DHT) (FC=-2.1234, p = 0.0165) between GDM and no-GDM. Stepwise logistic regression analysis resulted in the best predictive model, including six variables (Δweight, ΔF, Δcortisone (E), ΔE2, Δprogesterone (P), ΔDHT). The area under the curve for this model was 0.791, and for the external validation cohort, it was 0.799. CONCLUSIONS A GDM prediction model can be constructed using SH measures during early and mid-Down syndrome screening.
Collapse
Affiliation(s)
- Bo Zhu
- Department of Laboratory Medicine, The Women's Hospital of Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou, Zhejiang, China.
| | - Binbin Yin
- Department of Laboratory Medicine, The Women's Hospital of Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou, Zhejiang, China.
| | - Hui Li
- Department of Laboratory Medicine, The Women's Hospital of Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou, Zhejiang, China.
| | - Xuelian Chu
- Department of Laboratory Medicine, Hangzhou Linping District Maternal and Child Health Hospital, 359 Renmin Road, Hangzhou, Zhejiang, China.
| | - Zhifeng Mi
- Department of Laboratory Medicine, Haining Maternal and Child Healthcare Hospital, 309 Shui Yue Ting East Road, Jiaxing, Zhejiang, China.
| | - Yanni Sun
- Department of Laboratory Medicine, The Women's Hospital of Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou, Zhejiang, China.
| | - Xiaofen Yuan
- Hangzhou Calibra Diagnostics Co., Ltd, Gene Town, Zijin Park, 859 Shixiang West Road, Hangzhou, Zhejiang, China.
| | - Rongchang Chen
- Hangzhou Calibra Diagnostics Co., Ltd, Gene Town, Zijin Park, 859 Shixiang West Road, Hangzhou, Zhejiang, China.
| | - Zhixin Ma
- Department of Laboratory Medicine, The Women's Hospital of Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou, Zhejiang, China.
| |
Collapse
|
29
|
Ammar L, Bird K, Nian H, Maxwell-Horn A, Lee R, Ding T, Riddell C, Gebretsadik T, Snyder B, Hartert T, Wu P. Development and Validation of a Diagnostic Algorithm for Down Syndrome Using Birth Certificate and International Classification of Diseases Codes. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1271. [PMID: 39457236 PMCID: PMC11506645 DOI: 10.3390/children11101271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVE We aimed to develop an algorithm that accurately identifies children with Down syndrome (DS) using administrative data. METHODS We identified a cohort of children born between 2000 and 2017, enrolled in the Tennessee Medicaid Program (TennCare), who either had DS coded on their birth certificate or had a diagnosis listed using an International Classification of Diseases (ICD) code (suspected DS), and who received care at Vanderbilt University Medical Center, a comprehensive academic medical center, in the United States. Children with suspected DS were defined as having DS if they had (a) karyotype-confirmed DS indicated on their birth certificate; (b) karyotype-pending DS indicated on their birth certificate (or just DS if test type was not specified) and at least two healthcare encounters for DS during the first 6 years of life; or (c) at least three healthcare encounters for DS, with the first and last encounter separated by at least 30 days, during the first six years of life. The positive predictive value (PPV) of the algorithm and 95% confidence interval (CI) were reported. RESULTS Of the 411 children with suspected DS, 354 (86.1%) were defined as having DS by the algorithm. According to medical chart review, the algorithm correctly identified 347 children with DS (PPV = 98%, 95%CI: 96.0-99.0%). Of the 57 children the algorithm defined as not having DS, 50 (97.7%, 95%CI: 76.8-93.9%) were confirmed as not having DS by medical chart review. CONCLUSIONS An algorithm that accurately identifies individuals with DS using birth certificate data and/or ICD codes provides a valuable tool to study DS using administrative data.
Collapse
Affiliation(s)
- Lin Ammar
- Division of Epidemiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
| | - Kristin Bird
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (K.B.); (A.M.-H.); (T.H.)
| | - Hui Nian
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (H.N.); (T.D.); (T.G.)
| | - Angela Maxwell-Horn
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (K.B.); (A.M.-H.); (T.H.)
| | - Rees Lee
- Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85721, USA;
| | - Tan Ding
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (H.N.); (T.D.); (T.G.)
| | - Corinne Riddell
- Division of Biostatistics and Epidemiology, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Tebeb Gebretsadik
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (H.N.); (T.D.); (T.G.)
| | - Brittney Snyder
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Tina Hartert
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (K.B.); (A.M.-H.); (T.H.)
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Pingsheng Wu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (H.N.); (T.D.); (T.G.)
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| |
Collapse
|
30
|
Szczawińska-Popłonyk A, Popłonyk N, Awdi K. Down Syndrome in Children: A Primary Immunodeficiency with Immune Dysregulation. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1251. [PMID: 39457216 PMCID: PMC11506678 DOI: 10.3390/children11101251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/06/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024]
Abstract
Background: The multisystemic features of Down syndrome (DS) in children are accompanied by immunodeficiency, making them susceptible to infections and immune dysregulation with autoimmune, allergic, inflammatory, and hematological complications. This study was aimed at a better understanding of the abnormalities within the B and T cell compartments and their correlations with clinical immunophenotypes. Methods: Medical records of 35 DS children were retrospectively reviewed, referring to clinical symptomatology including history of infections, immune dysregulation disorders, and humoral and cellular immune response. Results: While the etiology of respiratory tract infections included typical viral and bacterial pathogens, SARS-CoV2-induced inflammatory disease and syndromic immunodeficiency contributed significantly to the deterioration of the clinical course. Allergic diseases in the form of asthma, allergic rhinitis, and alimentary allergy were the most frequent manifestations of immune dysregulation and were followed by autoimmune disorders, such as Crohn's disease, celiac disease, autoimmune thyroiditis, and alopecia, as well as inflammatory disorders, balanitis xerotica obliterans and lymphadenopathy, and a hematological disorder of myelopoiesis. Deficiency of serum immunoglobulin levels, reduced numbers of naïve B cells, and non-switched memory B cells along with low naïve T helper cells and significantly reduced regulatory T helper cells were the most prominent immune abnormalities. Conclusions: The loss of naïveté in B and T lymphocyte compartments with a deficiency of regulatory T cells may be underpinning pathomechanisms for the skewed immune response. The clinical immunophenotype in DS is complex and represents syndromic primary immunodeficiency with immune dysregulation.
Collapse
Affiliation(s)
- Aleksandra Szczawińska-Popłonyk
- Department of Pediatric Pneumonology, Allergy and Clinical Immunology, Institute of Pediatrics, Poznań University of Medical Sciences, Szpitalna 27/33, 60-572 Poznań, Poland
| | - Natalia Popłonyk
- Student Scientific Society, Poznań University of Medical Sciences, 61-701 Poznań, Poland
| | - Karina Awdi
- Student Scientific Society, English Division, Poznań University of Medical Sciences, 61-701 Poznań, Poland
| |
Collapse
|
31
|
Wohlfert AJ, Phares J, Granholm AC. The mTOR Pathway: A Common Link Between Alzheimer's Disease and Down Syndrome. J Clin Med 2024; 13:6183. [PMID: 39458132 PMCID: PMC11508835 DOI: 10.3390/jcm13206183] [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: 09/04/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Down syndrome (DS) is a chromosomal condition that causes many systemic dysregulations, leading to several possible age-related diseases including Alzheimer's disease (AD). This may be due to the triplication of the Amyloid precursor protein (APP) gene or other alterations in mechanistic pathways, such as the mTOR pathway. Impairments to upstream regulators of mTOR, such as insulin, PI3K/AKT, AMPK, and amino acid signaling, have been linked to amyloid beta plaques (Aβ) and neurofibrillary tangles (NFT), the most common AD pathologies. However, the mechanisms involved in the progression of pathology in human DS-related AD (DS-AD) are not fully investigated to date. Recent advancements in omics platforms are uncovering new insights into neurodegeneration. Genomics, spatial transcriptomics, proteomics, and metabolomics are novel methodologies that provide more data in greater detail than ever before; however, these methods have not been used to analyze the mTOR pathways in connection to DS-AD. Using these new techniques can unveil unexpected insights into pathological cellular mechanisms through an unbiased approach.
Collapse
Affiliation(s)
- Abigail J. Wohlfert
- Department of Modern Human Anatomy and Cell & Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Jeremiah Phares
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| | - Ann-Charlotte Granholm
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| |
Collapse
|
32
|
Riha RL, Singh A, Hill EA, Evans H, O'Regan D. Sleep-disordered breathing in children and adults with intellectual disability: mind the gap! Thorax 2024; 79:1099-1107. [PMID: 38937106 DOI: 10.1136/thorax-2023-220032] [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: 08/03/2023] [Accepted: 05/08/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND In adults and children with intellectual disability (ID), sleep -disordered breathing (SDB) is thought to be common. However, large epidemiological studies are lacking, and there are few studies on optimal methods of investigation and even fewer randomised, controlled intervention trials of treatment. METHOD Peer-reviewed publications from various databases were examined in line with search terms relevant to ID and SDB spanning the years 200-2024. RESULTS Findings suggest that, due to comorbid conditions, children and adults with ID may experience both an increased risk of SDB, as well as lower frequency of diagnosis. SDB can compromise the emotional, physical and mental health of individuals with ID. Appropriate treatment when tolerated leads to an improvement in health and well-being and several studies emphasized the importance of consistent follow-up of people with ID - something that is not universally occurring during childhood, in the transition to adulthood and during adulthood itself. As the most frequently occurring form of ID worldwide, we use Down syndrome as a specific example of how diagnosing and treating SDB can lead to improved outcomes. CONCLUSIONS This review highlights the importance of identifying SDB in this heterogenous population, recognising the multi-faceted, deleterious consequences of untreated SDB in people with ID, and presents some strategies that can be harnessed to improve diagnosis and management. Until further ID-specific research is available, we urge flexibility in the approach to people with ID and SDB based in guidelines and standard practice developed for the typically developing population.
Collapse
Affiliation(s)
- Renata L Riha
- Department of Sleep Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Ankur Singh
- Department of Sleep Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Elizabeth A Hill
- School of Applied Sciences, College of Health, Science and Society, University of the West of England, Bristol, UK
| | - Hazel Evans
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - David O'Regan
- Sleep Disorders Centre, Guy's and St Thomas' Hospital NHS Foundation Trust; Faculty of Life and Sciences Medicine, King's College London, London, UK
| |
Collapse
|
33
|
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
|
34
|
Rojas J, Hose J, Dutcher HA, Place M, Wolters JF, Hittinger CT, Gasch AP. Comparative modeling reveals the molecular determinants of aneuploidy fitness cost in a wild yeast model. CELL GENOMICS 2024; 4:100656. [PMID: 39317188 PMCID: PMC11602619 DOI: 10.1016/j.xgen.2024.100656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/10/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024]
Abstract
Although implicated as deleterious in many organisms, aneuploidy can underlie rapid phenotypic evolution. However, aneuploidy will be maintained only if the benefit outweighs the cost, which remains incompletely understood. To quantify this cost and the molecular determinants behind it, we generated a panel of chromosome duplications in Saccharomyces cerevisiae and applied comparative modeling and molecular validation to understand aneuploidy toxicity. We show that 74%-94% of the variance in aneuploid strains' growth rates is explained by the cumulative cost of genes on each chromosome, measured for single-gene duplications using a genomic library, along with the deleterious contribution of small nucleolar RNAs (snoRNAs) and beneficial effects of tRNAs. Machine learning to identify properties of detrimental gene duplicates provided no support for the balance hypothesis of aneuploidy toxicity and instead identified gene length as the best predictor of toxicity. Our results present a generalized framework for the cost of aneuploidy with implications for disease biology and evolution.
Collapse
Affiliation(s)
- Julie Rojas
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - James Hose
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - H Auguste Dutcher
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael Place
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA; Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - John F Wolters
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chris Todd Hittinger
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA; Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA; J.F. Crow Institute for the Study of Evolution, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Audrey P Gasch
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA; Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA; J.F. Crow Institute for the Study of Evolution, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
35
|
Wang Y, Fu X, Shen Y. The hidden costs of aneuploidy: New insights from yeast. CELL GENOMICS 2024; 4:100673. [PMID: 39389013 PMCID: PMC11602565 DOI: 10.1016/j.xgen.2024.100673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024]
Abstract
The molecular mechanisms underlying the paradoxical effects1 of aneuploidy are still not completely understood. In this issue, Rojas et al.2 systematically analyzed the associated costs of aneuploidy and the molecular drivers involved, which revealed that aneuploidy stress is primarily driven by the cumulative effects of genes per chromosome. Notably, gene length was predicted as the most significant indicator of aneuploidy toxicity by machine learning.
Collapse
Affiliation(s)
- Yuerong Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI Research, Changzhou 213299, China; BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518083, China
| | - Xian Fu
- BGI Research, Changzhou 213299, China; BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518083, China
| | - Yue Shen
- BGI Research, Changzhou 213299, China; BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518083, China.
| |
Collapse
|
36
|
Yang J, Zhao W, Liang L, Chi G. Treatment of iliac arteriovenous malformation associated with trisomy 21: a case report. J Cardiothorac Surg 2024; 19:594. [PMID: 39375784 PMCID: PMC11457334 DOI: 10.1186/s13019-024-03062-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 09/15/2024] [Indexed: 10/09/2024] Open
Abstract
Both arteriovenous malformation (AVM) and trisomy 21 are rare diseases. Studies have shown that individuals with trisomy 21 may have potential vascular malformations, with the main site of onset being the portal system. This case involved an adult patient with trisomy 21 who presented with pelvic iliac arteriovenous malformations. The patient underwent interventional treatment, and the prognosis was favorable. It is important to enhance the screening for vascular malformations in patients with trisomy 21 syndrome. At the same time, interventional treatment for AVMs is effective and increasingly important for managing AVMs.
Collapse
Affiliation(s)
- Jinming Yang
- Department of Vascular Interventional, Aerospace Center Hospital, No. 15 Yuquan Road, Haidian District, Beijing, 100049, People's Republic of China
| | - Weichao Zhao
- Department of Surgery, Nanfen Hospital of Benxi Iron & Steel Industry Group of Liaoning Health Industry Group, No. 30 Nanshan Road, Nanfen District, Benxi, 117014, Liaoning Province, People's Republic of China
| | - Liping Liang
- Department of Interventional Surgery, Aerospace Center Hospital, No. 15 Yuquan Road, Haidian District, Beijing, 100049, People's Republic of China
| | - Guoqing Chi
- Department of Vascular Interventional, Aerospace Center Hospital, No. 15 Yuquan Road, Haidian District, Beijing, 100049, People's Republic of China.
| |
Collapse
|
37
|
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
|
38
|
Dutcher HA, Hose J, Howe H, Rojas J, Gasch AP. The response to single-gene duplication implicates translation as a key vulnerability in aneuploid yeast. PLoS Genet 2024; 20:e1011454. [PMID: 39453980 PMCID: PMC11540229 DOI: 10.1371/journal.pgen.1011454] [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: 04/22/2024] [Revised: 11/06/2024] [Accepted: 10/07/2024] [Indexed: 10/27/2024] Open
Abstract
Aneuploidy produces myriad consequences in health and disease, yet models of the deleterious effects of chromosome amplification are still widely debated. To distinguish the molecular determinants of aneuploidy stress, we measured the effects of duplicating individual genes in cells with different chromosome duplications, in wild-type cells (SSD1+) and cells sensitized to aneuploidy by deletion of RNA-binding protein Ssd1 (ssd1Δ). We identified gene duplications that are nearly neutral in wild-type euploid cells but significantly deleterious in euploids lacking SSD1 or in SSD1+ aneuploid cells with different chromosome duplications. Several of the most deleterious genes are linked to translation. In contrast, duplication of other genes benefits multiple ssd1Δ aneuploids over controls, and this group is enriched for translational effectors. Furthermore, both wild-type and especially ssd1Δ aneuploids with different chromosome amplifications show increased sensitivity to translational inhibitor nourseothricin. We used comparative modeling of aneuploid growth defects, based on the cumulative fitness costs measured for single-gene duplication. Our results present a model in which the deleterious effects of aneuploidy emerge from an interaction between the cumulative burden of many amplified genes on a chromosome and a subset of duplicated genes that become toxic in that context. These findings provide a perspective on the dual impact of individual genes and overall genomic burden, offering new avenues for understanding aneuploidy and its cellular consequences.
Collapse
Affiliation(s)
- H. Auguste Dutcher
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - James Hose
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Hollis Howe
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Julie Rojas
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Audrey P. Gasch
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
39
|
Malinge S. Childhood leukaemia in Down's syndrome primed by blood-cell bias. Nature 2024; 634:40-42. [PMID: 39322696 DOI: 10.1038/d41586-024-02785-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
|
40
|
Talukder A, Yeung D, Li Y, Anandanadarajah N, Umbach DM, Fan Z, Li L. Comparison of power spectra from overnight electroencephalography between patients with Down syndrome and matched control subjects. J Sleep Res 2024; 33:e14187. [PMID: 38410055 PMCID: PMC11347723 DOI: 10.1111/jsr.14187] [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: 10/02/2023] [Revised: 01/31/2024] [Accepted: 02/16/2024] [Indexed: 02/28/2024]
Abstract
Electroencephalograms can capture brain oscillatory activities during sleep as a form of electrophysiological signals. We analysed electroencephalogram recordings from full-night in-laboratory polysomnography from 100 patients with Down syndrome, and 100 age- and sex-matched controls. The ages of patients with Down syndrome spanned 1 month to 31 years (median 4.4 years); 84 were younger than 12 years, and 54 were male. From each electroencephalogram, we extracted relative power in six frequency bands or rhythms (delta, theta, alpha, slow sigma, fast sigma, and beta) from six channels (frontal F3 and F4, central C3 and C4, and occipital O1 and O2) during five sleep stages (N3, N2, N1, R and W)-180 features in all. We examined differences in relative power between Down syndrome and control electroencephalograms for each feature separately. During wake and N1 sleep stages, alpha rhythms (8.0-10.5 Hz) had significantly lower power in patients with Down syndrome than controls. Moreover, the rate of increase in alpha power with age during rapid eye movement sleep was significantly slower in Down syndrome than control subjects. During wake and N1 sleep, delta rhythms (0.25-4.5 Hz) had higher power in patients with Down syndrome than controls. During N2 sleep, slow sigma rhythms (10.5-12.5 Hz) had lower power in patients with DS than controls. These findings extend previous research from routine electroencephalogram studies demonstrating that patients with Down syndrome had reduced circadian amplitude-the difference between wake alpha power and deep sleep delta power was smaller in Down syndrome than control subjects. We envision that these brain oscillatory activities may be used as surrogate markers for clinical trials for patients with Down syndrome.
Collapse
Affiliation(s)
- Amlan Talukder
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Deryck Yeung
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Yuanyuan Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Nishanth Anandanadarajah
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - David M. Umbach
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Zheng Fan
- Division of Sleep Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Leping Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| |
Collapse
|
41
|
Chachlaki K, Duc KL, Storme L, Prévot V. Novel insights into minipuberty and GnRH: Implications on neurodevelopment, cognition, and COVID-19 therapeutics. J Neuroendocrinol 2024; 36:e13387. [PMID: 38565500 PMCID: PMC7616535 DOI: 10.1111/jne.13387] [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: 01/03/2024] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
In humans, the first 1000 days of life are pivotal for brain and organism development. Shortly after birth, gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus are activated, a phenomenon known as minipuberty. This phenomenon, observed in all mammals studied, influences the postnatal development of the hypothalamic-pituitary-gonadal (HPG) axis and reproductive function. This review will put into perspective the results of recent studies showing that the impact of minipuberty extends beyond reproductive function, influencing sensory and cognitive maturation. Studies in mice have revealed the role of nitric oxide (NO) in regulating minipuberty amplitude, with NO deficiency linked to cognitive and olfactory deficits. Additionally, findings indicate that cognitive and sensory defects in adulthood in a mouse model of Down syndrome are associated with an age-dependent decline of GnRH production, whose origin can be traced back to minipuberty, and point to the potential therapeutic role of pulsatile GnRH administration in cognitive disorders. Furthermore, this review delves into the repercussions of COVID-19 on GnRH production, emphasizing potential consequences for neurodevelopment and cognitive function in infected individuals. Notably, GnRH neurons appear susceptible to SARS-CoV-2 infection, raising concerns about potential long-term effects on brain development and function. In conclusion, the intricate interplay between GnRH neurons, GnRH release, and the activity of various extrahypothalamic brain circuits reveals an unexpected role for these neuroendocrine neurons in the development and maintenance of sensory and cognitive functions, supplementing their established function in reproduction. Therapeutic interventions targeting the HPG axis, such as inhaled NO therapy in infancy and pulsatile GnRH administration in adults, emerge as promising approaches for addressing neurodevelopmental cognitive disorders and pathological aging.
Collapse
Affiliation(s)
- Konstantina Chachlaki
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
| | - Kevin Le Duc
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
- CHU Lille, Neonatology Department, Jeanne de Flandres Hospital, Lille, France
| | - Laurent Storme
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
- CHU Lille, Neonatology Department, Jeanne de Flandres Hospital, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
| |
Collapse
|
42
|
Chang P, Pérez-González M, Constable J, Bush D, Cleverley K, Tybulewicz VLJ, Fisher EMC, Walker MC. Neuronal oscillations in cognition: Down syndrome as a model of mouse to human translation. Neuroscientist 2024:10738584241271414. [PMID: 39316548 DOI: 10.1177/10738584241271414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Down syndrome (DS), a prevalent cognitive disorder resulting from trisomy of human chromosome 21 (Hsa21), poses a significant global health concern. Affecting approximately 1 in 800 live births worldwide, DS is the leading genetic cause of intellectual disability and a major predisposing factor for early-onset Alzheimer's dementia. The estimated global population of individuals with DS is 6 million, with increasing prevalence due to advances in DS health care. Global efforts are dedicated to unraveling the mechanisms behind the varied clinical outcomes in DS. Recent studies on DS mouse models reveal disrupted neuronal circuits, providing insights into DS pathologies. Yet, translating these findings to humans faces challenges due to limited systematic electrophysiological analyses directly comparing human and mouse. Additionally, disparities in experimental procedures between the two species pose hurdles to successful translation. This review provides a concise overview of neuronal oscillations in human and rodent cognition. Focusing on recent DS mouse model studies, we highlight disruptions in associated brain function. We discuss various electrophysiological paradigms and suggest avenues for exploring molecular dysfunctions contributing to DS-related cognitive impairments. Deciphering neuronal oscillation intricacies holds promise for targeted therapies to alleviate cognitive disabilities in DS individuals.
Collapse
Affiliation(s)
- Pishan Chang
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London, UK
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK
| | | | - Jessica Constable
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London, UK
| | - Daniel Bush
- Department of Neuroscience, Physiology, and Pharmacology, UCL, London, UK
| | - Karen Cleverley
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London, UK
| | - Victor L J Tybulewicz
- Immune Cell Biology and Down Syndrome Laboratory, The Francis Crick Institute, London, UK
| | | | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| |
Collapse
|
43
|
Abed H, Taju W. Perspectives on managing patients with Down's syndrome among orthodontists in Saudi Arabia: A vignette and a COM-B analysis study. J Orthod Sci 2024; 13:33. [PMID: 39450221 PMCID: PMC11500740 DOI: 10.4103/jos.jos_14_24] [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: 02/01/2024] [Revised: 05/11/2024] [Accepted: 06/15/2024] [Indexed: 10/26/2024] Open
Abstract
OBJECTIVE This study aims to explore perspectives on managing patients with Down's syndrome among orthodontists in Saudi Arabia using a vignette study and COM-B model. METHODS This was a cross-sectional study for orthodontists working in Saudi Arabia. Participants were asked about their demographic data. Vignette (clinical scenarios) and COM-B model (C: capability, O: opportunity, M: motivation, and B: behavior) were used to assess orthodontists' perspectives regarding the management of patients with Down's syndrome. A model of Alpha (Cronbach) was used to study the properties of measurement scales. A Pearson's correlation coefficient was used to correlate variables represented by means. An independent t-test and one-way ANOVA, with least significant difference (LSD) as a post hoc test, were used to compare two group means and more than two groups, respectively. RESULTS Most participants (n = 70, 85.4%) reported that they had no training/exposure to dealing with Down's syndrome patients during their orthodontic clinical training. Participants reported an average of 3.82 (SD = 1.1) when treating moderate Down's syndrome and 2.48 (SD = 1.4) when treating severe Down's syndrome with mild occlusion discrepancies. Participants also showed that their behavior and attitude are significantly related to their capability, opportunity, and motivation regarding the management of patients with Down's syndrome. The average reported agreement ranges from 2.46 (SD = 1) for the hypoplasia of the mandible to 4.38 (SD = 0.8) for the congenitally missing teeth. CONCLUSION This study found that orthodontists in Saudi Arabia showed low confidence levels to manage Down's syndrome patients with moderate and severe challenging behavior. Despite this, the likelihood of increasing their confidence levels and attitude regarding the management of patients with Down's syndrome increased by increasing their capability, opportunity, motivation, and knowledge in this field.
Collapse
Affiliation(s)
- Hassan Abed
- Department of Basic and Clinical Oral Sciences, Faculty of Dentistry, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Waleed Taju
- Department of Preventive Dentistry, Faculty of Dentistry, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
44
|
Zhou X, Zeng X, Fang J, He J, Kuang H, Hua X, Wang A. Comparison of total prevalence, perinatal prevalence, and livebirth prevalence of birth defects in Hunan Province, China, 2016-2020. Front Public Health 2024; 12:1297426. [PMID: 39324160 PMCID: PMC11422065 DOI: 10.3389/fpubh.2024.1297426] [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: 09/20/2023] [Accepted: 07/08/2024] [Indexed: 09/27/2024] Open
Abstract
Objective Birth defect of any type is undesirable and often pose a negative impact on the health and development of the newborn. Birth defects surveillance with datasets from surveillance health-related programs are useful to predict the pattern of birth defects and take preventive measures. In this study, the total prevalence, perinatal prevalence, and livebirth prevalence of birth defects were compared. Methods Data were obtained from the Birth Defects Surveillance System in Hunan Province, China, 2016-2020. The total prevalence is the number of birth defects (including livebirths, stillbirths, and selective terminations of pregnancy) per 1,000 births (including livebirths and stillbirths). The perinatal prevalence is the number of birth defects (between 28 weeks gestation and 7 days postpartum) per 1,000 births. The livebirth prevalence is the number of liveborn birth defects per 1,000 births (unit: ‰). Underestimated proportion (unit: %) is the reduction level of perinatal prevalence or livebirth prevalence compared to the total prevalence. Prevalence with 95% confidence intervals (CI) was calculated using the log-binomial method. Chi-square tests (χ 2) were used to examine if significant differences existed in prevalence or underestimated proportion between different groups. Results A total of 847,755 births were included in this study, and 23,420 birth defects were identified, including 14,459 (61.74%) birth defects with gestational age > =28 weeks, and 11,465 (48.95%) birth defects in livebirths. The total prevalence, perinatal prevalence, and livebirth prevalence of birth defects were 27.63‰ (95%CI, 27.27-27.98), 17.06‰ (95%CI, 16.78-17.33), and 13.52‰ (95%CI, 13.28-13.77), respectively, and significant differences existed between them (χ2 = 4798.55, p < 0.01). Compared to the total prevalence, the perinatal prevalence and livebirth prevalence were underestimated by 38.26 and 51.05%, respectively. Significant differences existed between the total prevalence, perinatal prevalence, and livebirth prevalence of birth defects in all subgroups according to year, sex, residence, and maternal age (p < 0.05). Significant differences existed between the total prevalence, perinatal prevalence, and livebirth prevalence for 17 specific defects: congenital heart defect, cleft lip-palate, Down syndrome, talipes equinovarus, hydrocephalus, limb reduction, cleft lip, omphalocele, anal atresia, anencephaly, spina bifida, diaphragmatic hernia, encephalocele, gastroschisis, esophageal atresia, bladder exstrophy, and conjoined twins (p < 0.05). In comparison, no significant difference existed between the total prevalence, perinatal prevalence, and livebirth prevalence for 6 specific defects: polydactyly, other external ear defects, syndactyly, hypospadias, cleft palate, and anotia/microtia (p > 0.05). Conclusion The total prevalence and livebirth prevalence of birth defects in Hunan Province, China, was not well studied. A systematic study was conducted to compare the total prevalence, perinatal prevalence, and livebirth prevalence of birth defects. The study reveals that significant differences existed between the total prevalence, perinatal prevalence, and livebirth prevalence of birth defects (including many specific defects), and year, sex, residence, and maternal age had significant impacts on it. The outcomes of the study will help to take preventive measures for birth defects as well as benefit the people involving public health and policymakers to improve the current scenario.
Collapse
Affiliation(s)
- Xu Zhou
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Xiu Zeng
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Junqun Fang
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Jian He
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Haiyan Kuang
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Xinjun Hua
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Aihua Wang
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| |
Collapse
|
45
|
Otte ED, Roper RJ. Skeletal health in DYRK1A syndrome. Front Neurosci 2024; 18:1462893. [PMID: 39308945 PMCID: PMC11413744 DOI: 10.3389/fnins.2024.1462893] [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: 07/10/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
DYRK1A syndrome results from a reduction in copy number of the DYRK1A gene, which resides on human chromosome 21 (Hsa21). DYRK1A has been implicated in the development of cognitive phenotypes associated with many genetic disorders, including Down syndrome (DS) and Alzheimer's disease (AD). Additionally, overexpression of DYRK1A in DS has been implicated in the development of abnormal skeletal phenotypes in these individuals. Analyses of mouse models with Dyrk1a dosage imbalance (overexpression and underexpression) show skeletal deficits and abnormalities. Normalization of Dyrk1a copy number in an otherwise trisomic animal rescues some skeletal health parameters, and reduction of Dyrk1a copy number in an otherwise euploid (control) animal results in altered skeletal health measurements, including reduced bone mineral density (BMD) in the femur, mandible, and skull. However, little research has been conducted thus far on the implications of DYRK1A reduction on human skeletal health, specifically in individuals with DYRK1A syndrome. This review highlights the skeletal phenotypes of individuals with DYRK1A syndrome, as well as in murine models with reduced Dyrk1a copy number, and provides potential pathways altered by a reduction of DYRK1A copy number, which may impact skeletal health and phenotypes in these individuals. Understanding how decreased expression of DYRK1A in individuals with DYRK1A syndrome impacts bone health may increase awareness of skeletal traits and assist in the development of therapies to improve quality of life for these individuals.
Collapse
Affiliation(s)
- Elysabeth D Otte
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, United States
| | - Randall J Roper
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, United States
| |
Collapse
|
46
|
Kozlov G, Franceschi C, Vedunova M. Intricacies of aging and Down syndrome. Neurosci Biobehav Rev 2024; 164:105794. [PMID: 38971514 DOI: 10.1016/j.neubiorev.2024.105794] [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: 04/11/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Down syndrome is the most frequently occurring genetic condition, with a substantial escalation in risk associated with advanced maternal age. The syndrome is characterized by a diverse range of phenotypes, affecting to some extent all levels of organization, and its progeroid nature - early manifestation of aspects of the senile phenotype. Despite extensive investigations, many aspects and mechanisms of the disease remain unexplored. The current review aims to provide an overview of the main causes and manifestations of Down syndrome, while also examining the phenomenon of accelerated aging and exploring potential therapeutic strategies.
Collapse
Affiliation(s)
- G Kozlov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Gagarin ave., 23, 603022, Russia
| | - C Franceschi
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Gagarin ave., 23, 603022, Russia
| | - M Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Gagarin ave., 23, 603022, Russia; Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| |
Collapse
|
47
|
Batbold S, Cummings G, Riggan KA, Michie M, Allyse M. Views of parents of children with Down syndrome on Alzheimer's disease vaccination. JOURNAL OF INTELLECTUAL DISABILITIES : JOID 2024; 28:759-772. [PMID: 37226465 DOI: 10.1177/17446295231177787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Individuals with Down syndrome (DS) experience increased risk of Alzheimer's disease (AD). Recent studies suggest that a vaccine against AD may be forthcoming. Parental buy-in is critical to the success of any intervention in this population, as adults with DS often rely on familial support. This study aims to characterize parents' perceptions of a hypothetical vaccine to prevent AD in individuals with DS. A mixed-methods, anonymous survey was distributed via social media. Participants were asked about their experiences with DS and reactions to proposed interventions. Open-ended responses were thematically analyzed using NVivo 12. Of 1,093 surveys initiated, 532 were completed. Of the parents sampled (N = 532), a small majority (54.3%), supported the proposed AD vaccine. All expressed the need for extensive pre-enrollment education and minimal risk. For many, limited research and long-term sequelae were concerns.
Collapse
Affiliation(s)
- Sarah Batbold
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Kirsten A Riggan
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN, USA
| | - Marsha Michie
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Megan Allyse
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
48
|
Yu H, Liu Y, Xu F, Fu Y, Yang M, Ding L, Wu Y, Tang F, Qiao J, Wen L. A human fetal cerebellar map of the late second trimester reveals developmental molecular characteristics and abnormality in trisomy 21. Cell Rep 2024; 43:114586. [PMID: 39137113 DOI: 10.1016/j.celrep.2024.114586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/29/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
Our understanding of human fetal cerebellum development during the late second trimester, a critical period for the generation of astrocytes, oligodendrocytes, and unipolar brush cells (UBCs), remains limited. Here, we performed single-cell RNA sequencing (scRNA-seq) in human fetal cerebellum samples from gestational weeks (GWs) 18-25. We find that proliferating UBC progenitors distribute in the subventricular zone of the rhombic lip (RLSVZ) near white matter (WM), forming a layer structure. We also delineate two trajectories from astrogenic radial glia (ARGs) to Bergmann glial progenitors (BGPs) and recognize oligodendrogenic radial glia (ORGs) as one source of primitive oligodendrocyte progenitor cells (PriOPCs). Additionally, our scRNA-seq analysis of the trisomy 21 fetal cerebellum at this stage reveals abnormal upregulated genes in pathways such as the cell adhesion pathway and focal adhesion pathway, which potentially promote neuronal differentiation. Overall, our research provides valuable insights into normal and abnormal development of the human fetal cerebellum.
Collapse
Affiliation(s)
- Hongmin Yu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
| | - Yun Liu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Changping Laboratory, Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China
| | - Fanqing Xu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Yuanyuan Fu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
| | - Ming Yang
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Ling Ding
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Yixuan Wu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Changping Laboratory, Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China
| | - Jie Qiao
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China.
| | - Lu Wen
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Changping Laboratory, Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China.
| |
Collapse
|
49
|
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
|
50
|
Amir M, Shafi S, Parveen S, Reshi AA, Ahmad A. Network Pharmacology Identifies Intersection Genes of Apigenin and Naringenin in Down Syndrome as Potential Therapeutic Targets. Pharmaceuticals (Basel) 2024; 17:1090. [PMID: 39204195 PMCID: PMC11359399 DOI: 10.3390/ph17081090] [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: 07/11/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Down Syndrome (DS), characterized by trisomy of chromosome 21, leads to the overexpression of several genes contributing to various pathologies, including cognitive deficits and early-onset Alzheimer's disease. This study aimed to identify the intersection genes of two polyphenolic compounds, apigenin and naringenin, and their potential therapeutic targets in DS using network pharmacology. Key proteins implicated in DS, comprising DYRK1A, APP, CBS, and ETS2, were selected for molecular docking and dynamics simulations to assess the binding affinities and stability of the protein-ligand interactions. Molecular docking revealed that naringenin exhibited the highest binding affinity to DYRK1A with a score of -9.3 kcal/mol, followed by CBS, APP, and ETS2. Moreover, molecular docking studies included positive control drugs, such as lamellarin D, valiltramiprosate, benserazide, and TK216, which exhibited binding affinities ranging from -5.5 to -8.9 kcal/mol. Apigenin showed strong binding to APP with a score of -8.8 kcal/mol, suggesting its potential in modulating amyloid-beta levels. These interactions were further validated through molecular dynamics simulations, demonstrating stable binding throughout the 100 ns simulation period. Root mean square deviation (RMSD) and root mean square fluctuation (RMSF) analyses indicated minimal fluctuations, confirming the stability of the complexes. The findings suggest that apigenin and naringenin could serve as effective therapeutic agents for DS by targeting key proteins involved in its pathology. Future studies should focus on in vivo validation, clinical trials, and exploring combination therapies to fully harness the therapeutic potential of these compounds for managing DS. This study underscores the promising role of network pharmacology in identifying novel therapeutic targets and agents for complex disorders like DS.
Collapse
Affiliation(s)
- Mohd Amir
- Department of Natural Products, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Shabana Shafi
- Department of Computer Science, College of Computer Science and Engineering, Taibah University, Madinah 42353, Saudi Arabia;
| | - Shahida Parveen
- Department of Nursing, College of Pharmacy and Applied Medical Sciences (CPAMS), Dar Al Uloom University, Riyadh 13314, Saudi Arabia;
| | - Aijaz Ahmad Reshi
- Department of Computer Science, College of Computer Science and Engineering, Taibah University, Madinah 42353, Saudi Arabia;
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|