1
|
Previdi A, Jordan P, Egloff C, Coussement A, Ahmed-Eli S, Tudal L, Bienvenu T, Picone O, Dupont JM. Prenatal diagnosis of a 15q24.1 microdeletion in a fetus with cerebral and urogenital abnormalities. Clin Genet 2024; 106:537-544. [PMID: 39012202 DOI: 10.1111/cge.14592] [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: 04/12/2024] [Revised: 06/21/2024] [Accepted: 07/05/2024] [Indexed: 07/17/2024]
Abstract
15q24.1 microdeletion syndrome is a recently described condition often resulting from non-allelic homologous recombination (NAHR). Typical clinical features include pre and post-natal growth retardation, facial dysmorphism, developmental delay and intellectual disability. Nonspecific urogenital, skeletal, and digit abnormalities may be present, although other congenital malformations are less frequent. Consequently, only one case was reported prenatally, complicating the genotype-phenotype correlation and the genetic counseling. We identified prenatally a second case, presenting with cerebral abnormalities including hydrocephaly, macrocephaly, cerebellum hypoplasia, vermis hypoplasia, rhombencephalosynapsis, right kidney agenesis with left kidney duplication and micropenis. Genome-wide aCGH assay allowed a diagnosis at 26 weeks of amenorrhea revealing a 1.6 Mb interstitial deletion on the long arm of chromosome 15 at 15q24.1-q24.2 (arr[GRCh37] 15q24.1q24.2(74,399,112_76,019,966)x1). A deep review of the literature was undertaken to further delineate the prenatal clinical features and the candidate genes involved in the phenotype. Cerebral malformations are typically nonspecific, but microcephaly appears to be the most frequent in postnatal cases. Our case is the first reported with a frank cerebellar involvement.
Collapse
Affiliation(s)
- Anaïk Previdi
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Pénélope Jordan
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Charles Egloff
- AP-HP.Nord-Université Paris Cité, Site Hôpital Louis Mourier, Service de Gynécologie Obstétrique, Colombes, France
| | - Aurélie Coussement
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Samira Ahmed-Eli
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Laure Tudal
- AP-HP.Nord-Université Paris Cité, Site Hôpital Louis Mourier, Service de Gynécologie Obstétrique, Colombes, France
| | - Thierry Bienvenu
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Olivier Picone
- AP-HP.Nord-Université Paris Cité, Site Hôpital Louis Mourier, Service de Gynécologie Obstétrique, Colombes, France
| | - Jean-Michel Dupont
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| |
Collapse
|
2
|
Zhang C, Wei G, Zhou H, Liu L. Causal relationships of familial hypercholesterolemia with the risk of multiple vitamin deficiencies: a Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1401260. [PMID: 39502567 PMCID: PMC11534809 DOI: 10.3389/fendo.2024.1401260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024] Open
Abstract
Background The causal relationship between familial hypercholesterolemia (FH) and various vitamin deficiencies has not yet been elucidated. Therefore, this study investigated the cause-and-effect relationship between FH and the risk of multiple vitamin deficiencies in humans. Methods Mendelian randomization (MR) analysis was performed by extracting six datasets for FH, FH with ischemic heart disease (IHD), and vitamin deficiency (vitamin A, thiamine, other B-group vitamins, and vitamin D) from the FinnGen study, covering a total of 329,115; 316,290; 354,932; 354,949; 355,411 and 355,238 individuals, respectively. Results FH was suggestively associated with higher odds of thiamine deficiency [inverse variance weighted odds ratio (ORIVW) 95% confidence interval (CI): 1.62 (1.03, 2.55), P = 0.036] and vitamin D deficiencies [ORIVW CI: 1.35 (1.04, 1.75), P = 0.024], low-density lipoprotein receptor (LDLR) rs112898275 variant, rs11591147 and rs499883 in proprotein convertase subtilisin/kexin 9 (PCSK9), rs9644862 in cyclin-dependent kinase inhibitor 2 B antisense RNA1 (CDKN2B-AS1), and rs142834163 in dedicator of cytokinesis 6 (DOCK6) and rs115478735 in ABO blood group (ABO) strongly influenced the risk of thiamine deficiency, while the rs7412 variant in apolipoprotein E (APOE) mostly influenced the risk of vitamin D deficiency. FH with IHD was suggestively associated with higher odds of vitamin D deficiency (ORIVW, weighted median [WM][95%CI]: 1.31 [1.05, 1.64]; 1.47 [1.10, 1.97]) (P = 0.018; 0.010) without any single significant SNPs observed. Conclusion FH was positively associated with increased risks of thiamine and vitamin D deficiencies, revealing a prospective and unfortunate complication of FH.
Collapse
Affiliation(s)
- Cheng Zhang
- National Drug Clinical Trial Center, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Huan Zhou
- National Drug Clinical Trial Center, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lin Liu
- National Drug Clinical Trial Center, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
3
|
Carter SWD, Fee EL, Usuda H, Oguz G, Ramasamy A, Amin Z, Agnihotri B, Wei Q, Xiawen L, Takahashi T, Takahashi Y, Ikeda H, Kumagai Y, Saito Y, Saito M, Mattar C, Evans MI, Illanes SE, Jobe AH, Choolani M, Kemp MW. Antenatal steroids elicited neurodegenerative-associated transcriptional changes in the hippocampus of preterm fetal sheep independent of lung maturation. BMC Med 2024; 22:338. [PMID: 39183288 PMCID: PMC11346182 DOI: 10.1186/s12916-024-03542-5] [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: 05/12/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Antenatal steroid therapy for fetal lung maturation is routinely administered to women at risk of preterm delivery. There is strong evidence to demonstrate benefit from antenatal steroids in terms of survival and respiratory disease, notably in infants delivered at or below 32 weeks' gestation. However, dosing remains unoptimized and lung benefits are highly variable. Current treatment regimens generate high-concentration, pulsatile fetal steroid exposures now associated with increased risk of childhood neurodevelopmental diseases. We hypothesized that damage-associated changes in the fetal hippocampal transcriptome would be independent of preterm lung function. METHODS Date-mated ewes carrying a single fetus at 122 ± 2dGA (term = 150dGA) were randomized into 4 groups: (i) Saline Control Group, 4×2ml maternal saline intramuscular(IM) injections at 12hr intervals (n = 11); or (ii) Dex High Group, 2×12mg maternal IM dexamethasone phosphate injections at 12hr intervals followed by 2×2ml IM saline injections at 12hr intervals (n = 12; representing a clinical regimen used in Singapore); or (iii) Dex Low Group, 4×1.5mg maternal IM dexamethasone phosphate injections 12hr intervals (n = 12); or (iv) Beta-Acetate Group, 1×0.125mg/kg maternal IM betamethasone acetate injection followed by 3×2ml IM sterile normal saline injections 12hr intervals (n = 8). Lambs were surgically delivered 48hr after first maternal injection at 122-125dGA, ventilated for 30min to establish lung function, and euthanised for necropsy and tissue collection. RESULTS Preterm lambs from the Dex Low and Beta-Acetate Groups had statistically and biologically significant lung function improvements (measured by gas exchange, lung compliance). Compared to the Saline Control Group, hippocampal transcriptomic data identified 879 differentially significant expressed genes (at least 1.5-fold change and FDR < 5%) in the steroid-treated groups. Pulsatile dexamethasone-only exposed groups (Dex High and Dex Low) had three common positively enriched differentially expressed pathways related in part to neurodegeneration ("Prion Disease", "Alzheimer's Disease", "Arachidonic Acid metabolism"). Adverse changes were independent of respiratory function during ventilation. CONCLUSIONS Our data suggests that exposure to antenatal steroid therapy is an independent cause of damage- associated transcriptomic changes in the brain of preterm, fetal sheep. These data highlight an urgent need for careful reconsideration and balancing of how antenatal steroids are used, both for patient selection and dosing regimens.
Collapse
Affiliation(s)
- Sean W D Carter
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore.
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
| | - Haruo Usuda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Gokce Oguz
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome #02-01, Singapore, 138632, Republic of Singapore
| | - Adaikalavan Ramasamy
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome #02-01, Singapore, 138632, Republic of Singapore
| | - Zubair Amin
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Neonatology Khoo Teck Puat, National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Biswas Agnihotri
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Neonatology Khoo Teck Puat, National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Qin Wei
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Liu Xiawen
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Tsukasa Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuki Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Hideyuki Ikeda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yusaku Kumagai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuya Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Masatoshi Saito
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Citra Mattar
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Mark I Evans
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Fetal Medicine Foundation of America, New York, NY, USA
| | - Sebastián E Illanes
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Reproductive Biology Program, Center for Biomedical Research and Innovation, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Alan H Jobe
- Centre for Pulmonary Biology, Cincinnati Children's Hospital Medical Centre, Cincinnati, OH, USA
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Matthew W Kemp
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- Women and Infants Research Foundation, Perth, WA, Australia
| |
Collapse
|
4
|
Dang Do AN, Sleat DE, Campbell K, Johnson NL, Zheng H, Wassif CA, Dale RK, Porter FD. Cerebrospinal Fluid Protein Biomarker Discovery in CLN3. J Proteome Res 2023; 22:2493-2508. [PMID: 37338096 PMCID: PMC11095826 DOI: 10.1021/acs.jproteome.3c00199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Syndromic CLN3-Batten is a fatal, pediatric, neurodegenerative disease caused by variants in CLN3, which encodes the endolysosomal transmembrane CLN3 protein. No approved treatment for CLN3 is currently available. The protracted and asynchronous disease presentation complicates the evaluation of potential therapies using clinical disease progression parameters. Biomarkers as surrogates to measure the progression and effect of potential therapeutics are needed. We performed proteomic discovery studies using cerebrospinal fluid (CSF) samples from 28 CLN3-affected and 32 age-similar non-CLN3 individuals. Proximal extension assay (PEA) of 1467 proteins and untargeted data-dependent mass spectrometry [MS; MassIVE FTP server (ftp://MSV000090147@massive.ucsd.edu)] were used to generate orthogonal lists of protein marker candidates. At an adjusted p-value of <0.1 and threshold CLN3/non-CLN3 fold-change ratio of 1.5, PEA identified 54 and MS identified 233 candidate biomarkers. Some of these (NEFL, CHIT1) have been previously linked with other neurologic conditions. Others (CLPS, FAM217B, QRICH2, KRT16, ZNF333) appear to be novel. Both methods identified 25 candidate biomarkers, including CHIT1, NELL1, and ISLR2 which had absolute fold-change ratios >2. NELL1 and ISLR2 regulate axonal development in neurons and are intriguing new candidates for further investigation in CLN3. In addition to identifying candidate proteins for CLN3 research, this study provides a comparison of two large-scale proteomic discovery methods in CSF.
Collapse
Affiliation(s)
- An N. Dang Do
- Unit on Cellular Stress in Development and Diseases, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - David E. Sleat
- Center for Advanced Biotechnology and Medicine, Rutgers Biomedical Health Sciences, Piscataway, New Jersey 08854, United States
- Department of Biochemistry and Molecular Biology, Robert-Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Piscataway, New Jersey 08854, United States
| | - Kiersten Campbell
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nicholas L. Johnson
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Haiyan Zheng
- Center for Advanced Biotechnology and Medicine, Rutgers Biomedical Health Sciences, Piscataway, New Jersey 08854, United States
| | - Christopher A. Wassif
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Ryan K. Dale
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Forbes D. Porter
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
5
|
The Novel RXR Agonist MSU-42011 Differentially Regulates Gene Expression in Mammary Tumors of MMTV-Neu Mice. Int J Mol Sci 2023; 24:ijms24054298. [PMID: 36901727 PMCID: PMC10001983 DOI: 10.3390/ijms24054298] [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: 01/15/2023] [Revised: 02/15/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023] Open
Abstract
Retinoid X receptor (RXR) agonists, which activate the RXR nuclear receptor, are effective in multiple preclinical cancer models for both treatment and prevention. While RXR is the direct target of these compounds, the downstream changes in gene expression differ between compounds. RNA sequencing was used to elucidate the effects of the novel RXRα agonist MSU-42011 on the transcriptome in mammary tumors of HER2+ mouse mammary tumor virus (MMTV)-Neu mice. For comparison, mammary tumors treated with the FDA approved RXR agonist bexarotene were also analyzed. Each treatment differentially regulated cancer-relevant gene categories, including focal adhesion, extracellular matrix, and immune pathways. The most prominent genes altered by RXR agonists positively correlate with survival in breast cancer patients. While MSU-42011 and bexarotene act on many common pathways, these experiments highlight the differences in gene expression between these two RXR agonists. MSU-42011 targets immune regulatory and biosynthetic pathways, while bexarotene acts on several proteoglycan and matrix metalloproteinase pathways. Exploration of these differential effects on gene transcription may lead to an increased understanding of the complex biology behind RXR agonists and how the activities of this diverse class of compounds can be utilized to treat cancer.
Collapse
|
6
|
Nikolenko VN, Rizaeva NA, Oganesyan MV, Vekhova KA, Alyautdinova NAF, Balan SI, Karashaeva TA, Bolotskaya AA. Brain commissures and related pathologies. NEUROLOGY, NEUROPSYCHIATRY, PSYCHOSOMATICS 2022. [DOI: 10.14412/2074-2711-2022-6-73-79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- V. N. Nikolenko
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University); Lomonosov Moscow State University
| | - N. A. Rizaeva
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University); Lomonosov Moscow State University
| | - M. V. Oganesyan
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University); Lomonosov Moscow State University
| | - K. A. Vekhova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University)
| | | | | | - T. A. Karashaeva
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University)
| | - A. A. Bolotskaya
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University)
| |
Collapse
|
7
|
Yang J, Jia Z, Song X, Shi J, Wang X, Zhao X, He K. Proteomic and clinical biomarkers for acute mountain sickness in a longitudinal cohort. Commun Biol 2022; 5:548. [PMID: 35668171 PMCID: PMC9170681 DOI: 10.1038/s42003-022-03514-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/18/2022] [Indexed: 11/17/2022] Open
Abstract
Ascending to high-altitude by non-high-altitude natives is a well-suited model for studying acclimatization to extreme environments. Acute mountain sickness (AMS) is frequently experienced by visitors. The diagnosis of AMS mainly depends on a self-questionnaire, revealing the need for reliable biomarkers for AMS. Here, we profiled 22 AMS symptom phenotypes, 65 clinical indexes, and plasma proteomic profiles of AMS via a combination of proximity extension assay and multiple reaction monitoring of a longitudinal cohort of 53 individuals. We quantified 1069 proteins and validated 102 proteins. Via differential analysis, machine learning, and functional association analyses. We found and validated that RET played an important role in the pathogenesis of AMS. With high-accuracies (AUCs > 0.9) of XGBoost-based models, we prioritized ADAM15, PHGDH, and TRAF2 as protective, predictive, and diagnostic biomarkers, respectively. Our findings shed light on the precision medicine for AMS and the understanding of acclimatization to high-altitude environments.
Collapse
Affiliation(s)
- Jing Yang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China
| | - Zhilong Jia
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China.
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China.
- Center for Artificial Intelligence in Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.
| | - Xinyu Song
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China
- Center for Artificial Intelligence in Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Jinlong Shi
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China
| | - Xiaoreng Wang
- Laboratory of Radiation Injury Treatment, Medical Innovation Research Division, PLA General Hospital, Beijing, China
| | - Xiaojing Zhao
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Kunlun He
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China.
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China.
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
8
|
Parodi L, Myserlis EP, Chung J, Georgakis MK, Mayerhofer E, Henry J, Montgomery BE, Moy M, Xu H, Malik R, Langefeld CD, Dichgans M, Woo D, Rosand J, Anderson CD. Shared genetic background between SARS-CoV-2 infection and large artery stroke. Int J Stroke 2022; 17:17474930221095696. [PMID: 35403514 DOI: 10.1177/17474930221095696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
BACKGROUND AND AIMS Increased risk of stroke, particularly large artery stroke (LAS), has been observed in patients with COVID-19. The biological processes underlying the observed higher risk are still unknown. We explored the association between stroke subtypes and COVID-19 susceptibility to understand whether biological mechanisms specific to SARS-CoV-2 uptake/infection could be leading to excess stroke risk in this population. PATIENTS AND METHODS We constructed a polygenic risk score (PRS) of COVID-19 susceptibility and tested its association with stroke subtypes using individual- and summary-level genetic data (SiGN, MEGASTROKE). We generated co-expression networks of genes involved in SARS-CoV-2 uptake/infection (ACE2, TMPRSS2, BEST3, ISLR2 and ADAM17) based on existing tissue expression libraries. Gene-based association testing was performed using S-PrediXcan and VEGAS2. Permutation independence tests were performed to assess SARS-CoV-2-related gene enrichment in stroke and its subtypes. RESULTS Our PRS demonstrated an association between COVID-19 susceptibility and LAS in SiGN (OR = 1.05 per SD increase, 95% CI: (1.00, 1.10), p = 0.04) and MEGASTROKE (β = 0.510, 95% CI: (0.242, 0.779), FDR-p = 0.0019). The SARS-CoV-2 risk-related ISLR2 co-expression gene network was significantly associated with genetic risk of LAS in aorta, tibial arteries, and multiple brain regions (P < 0.05). CONCLUSION Presence of genetic correlation and significant pathway enrichment suggest that increases in LAS risk reported in COVID-19 patients may be intrinsic to the viral infection, rather than a more generalized response to severe illness.
Collapse
Affiliation(s)
- Livia Parodi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Evangelos Pavlos Myserlis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jaeyoon Chung
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Marios K Georgakis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ernst Mayerhofer
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan Henry
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bailey E Montgomery
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mandy Moy
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Huichun Xu
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rainer Malik
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Carl D Langefeld
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Daniel Woo
- Department of Neurology, University of Cincinnati, Cincinnati, OH, USA
| | - Jonathan Rosand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christopher D Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
9
|
Oh S, Park H, Zhang X. Hybrid Clustering of Single-Cell Gene Expression and Spatial Information via Integrated NMF and K-Means. Front Genet 2021; 12:763263. [PMID: 34819947 PMCID: PMC8606648 DOI: 10.3389/fgene.2021.763263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/13/2021] [Indexed: 11/13/2022] Open
Abstract
Advances in single cell transcriptomics have allowed us to study the identity of single cells. This has led to the discovery of new cell types and high resolution tissue maps of them. Technologies that measure multiple modalities of such data add more detail, but they also complicate data integration. We offer an integrated analysis of the spatial location and gene expression profiles of cells to determine their identity. We propose scHybridNMF (single-cell Hybrid Nonnegative Matrix Factorization), which performs cell type identification by combining sparse nonnegative matrix factorization (sparse NMF) with k-means clustering to cluster high-dimensional gene expression and low-dimensional location data. We show that, under multiple scenarios, including the cases where there is a small number of genes profiled and the location data is noisy, scHybridNMF outperforms sparse NMF, k-means, and an existing method that uses a hidden Markov random field to encode cell location and gene expression data for cell type identification.
Collapse
Affiliation(s)
- Sooyoun Oh
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Haesun Park
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Xiuwei Zhang
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
10
|
Wojtowicz WM, Vielmetter J, Fernandes RA, Siepe DH, Eastman CL, Chisholm GB, Cox S, Klock H, Anderson PW, Rue SM, Miller JJ, Glaser SM, Bragstad ML, Vance J, Lam AW, Lesley SA, Zinn K, Garcia KC. A Human IgSF Cell-Surface Interactome Reveals a Complex Network of Protein-Protein Interactions. Cell 2021; 182:1027-1043.e17. [PMID: 32822567 PMCID: PMC7440162 DOI: 10.1016/j.cell.2020.07.025] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/19/2020] [Accepted: 07/17/2020] [Indexed: 12/17/2022]
Abstract
Cell-surface protein-protein interactions (PPIs) mediate cell-cell communication, recognition, and responses. We executed an interactome screen of 564 human cell-surface and secreted proteins, most of which are immunoglobulin superfamily (IgSF) proteins, using a high-throughput, automated ELISA-based screening platform employing a pooled-protein strategy to test all 318,096 PPI combinations. Screen results, augmented by phylogenetic homology analysis, revealed ∼380 previously unreported PPIs. We validated a subset using surface plasmon resonance and cell binding assays. Observed PPIs reveal a large and complex network of interactions both within and across biological systems. We identified new PPIs for receptors with well-characterized ligands and binding partners for “orphan” receptors. New PPIs include proteins expressed on multiple cell types and involved in diverse processes including immune and nervous system development and function, differentiation/proliferation, metabolism, vascularization, and reproduction. These PPIs provide a resource for further biological investigation into their functional relevance and may offer new therapeutic drug targets. Human IgSF interactome reveals complex network of cell-surface protein interactions Phylogenetic homology analysis predicts protein-protein interactions ∼380 previously unknown protein-protein interactions identified Deorphanization of receptors and new binding partners for well-studied receptors
Collapse
Affiliation(s)
- Woj M Wojtowicz
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Jost Vielmetter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ricardo A Fernandes
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dirk H Siepe
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Catharine L Eastman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gregory B Chisholm
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sarah Cox
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Heath Klock
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Paul W Anderson
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Sarah M Rue
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Jessica J Miller
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Scott M Glaser
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Melisa L Bragstad
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Julie Vance
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Annie W Lam
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Scott A Lesley
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
11
|
Melanotic Neuroectodermal Tumor of Infancy (MNTI) and Pineal Anlage Tumor (PAT) Harbor A Medulloblastoma Signature by DNA Methylation Profiling. Cancers (Basel) 2021; 13:cancers13040706. [PMID: 33572349 PMCID: PMC7916108 DOI: 10.3390/cancers13040706] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Melanotic neuroectodermal tumor of infancy (MNTI) is a rare tumor of uncertain origin, morphologically overlapping other rare neoplasms such as pineal anlage tumor (PAT) and a subset of medulloblastomas (i.e., melanotic medulloblastoma). Despite the similarities with MNTI, their possible histogenetic relationship has been traditionally disregarded based on their aggressive behavior and dismal prognosis. The aim of this study was to further characterize the molecular features of MNTI and PAT based on DNA-methylation and copy number variation profiling analysis. We found that MNTI shares a methylation profile with group 3 high-risk medulloblastoma, and potentially with PAT, suggesting a common histogenesis. Most MNTIs in our series lacked copy number variation alterations, whereas their presence in the one PAT deserves further study in larger cohorts to better determine their impact in prognosis and biologic behavior. Abstract MNTI is a rare tumor of indeterminate histogenesis and molecular signature. We performed methylation and copy number variation (CNV) profiles in patients with MNTI (n = 7) and PAT (n = 1) compared to the methylation brain tumor classifier v11b4 (BT-C) and the medulloblastoma (MB) classifier group 3/4 v1.0 (MB3/4-C). The patients’ mean age was 8 months (range: 4–48). The BT-C classified five MNTIs and one PAT (relapse) as class family MB-G3/G4, subclass group 3 (score: >0.9). The remaining two MNTIs and PAT (primary) were classified as class family plexus tumor, subclass pediatric (scores: >0.45). The MB3/4-C classified all MNTIs as high-risk MB-G3, Subtype II (score: >0.45). The primary PAT was classified as subtype III (score: 0.99) and its relapse as subtype II/III. MNTI and PAT clustered close to MB-G3. CNV analysis showed multiple rearrangements in one PAT and two MNTIs. The median follow-up was 54 months (four MNTIs in remission, one PAT died). In conclusion, we demonstrated that MNTI shares a homogenous methylation profile with MB-G3, and possibly with PAT. The role of a multipotent progenitor cell (i.e., early cranial neural crest cell) in their histogenesis and the influence of the anatomical site, tumor microenvironment, and other cytogenetic events in their divergent biologic behavior deserve further investigation.
Collapse
|
12
|
A gene expression atlas for different kinds of stress in the mouse brain. Sci Data 2020; 7:437. [PMID: 33328476 PMCID: PMC7744580 DOI: 10.1038/s41597-020-00772-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022] Open
Abstract
Stressful experiences are part of everyday life and animals have evolved physiological and behavioral responses aimed at coping with stress and maintaining homeostasis. However, repeated or intense stress can induce maladaptive reactions leading to behavioral disorders. Adaptations in the brain, mediated by changes in gene expression, have a crucial role in the stress response. Recent years have seen a tremendous increase in studies on the transcriptional effects of stress. The input raw data are freely available from public repositories and represent a wealth of information for further global and integrative retrospective analyses. We downloaded from the Sequence Read Archive 751 samples (SRA-experiments), from 18 independent BioProjects studying the effects of different stressors on the brain transcriptome in mice. We performed a massive bioinformatics re-analysis applying a single, standardized pipeline for computing differential gene expression. This data mining allowed the identification of novel candidate stress-related genes and specific signatures associated with different stress conditions. The large amount of computational results produced was systematized in the interactive “Stress Mice Portal”.
Collapse
|
13
|
Vijayalingam S, Ezekiel UR, Xu F, Subramanian T, Geerling E, Hoelscher B, San K, Ganapathy A, Pemberton K, Tycksen E, Pinto AK, Brien JD, Beck DB, Chung WK, Gurnett CA, Chinnadurai G. Human iPSC-Derived Neuronal Cells From CTBP1-Mutated Patients Reveal Altered Expression of Neurodevelopmental Gene Networks. Front Neurosci 2020; 14:562292. [PMID: 33192249 PMCID: PMC7653094 DOI: 10.3389/fnins.2020.562292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/01/2020] [Indexed: 11/17/2022] Open
Abstract
A recurrent de novo mutation in the transcriptional corepressor CTBP1 is associated with neurodevelopmental disabilities in children (Beck et al., 2016, 2019; Sommerville et al., 2017). All reported patients harbor a single recurrent de novo heterozygous missense mutation (p.R342W) within the cofactor recruitment domain of CtBP1. To investigate the transcriptional activity of the pathogenic CTBP1 mutant allele in physiologically relevant human cell models, we generated induced pluripotent stem cells (iPSC) from the dermal fibroblasts derived from patients and normal donors. The transcriptional profiles of the iPSC-derived “early” neurons were determined by RNA-sequencing. Comparison of the RNA-seq data of the neurons from patients and normal donors revealed down regulation of gene networks involved in neurodevelopment, synaptic adhesion and anti-viral (interferon) response. Consistent with the altered gene expression patterns, the patient-derived neurons exhibited morphological and electrophysiological abnormalities, and susceptibility to viral infection. Taken together, our studies using iPSC-derived neuron models provide novel insights into the pathological activities of the CTBP1 p.R342W allele.
Collapse
Affiliation(s)
- S Vijayalingam
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - Uthayashanker R Ezekiel
- Department of Clinical Health Sciences, Doisy College of Health Science, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Fenglian Xu
- Department of Biology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| | - T Subramanian
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - Brittany Hoelscher
- Department of Biology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| | - KayKay San
- Department of Clinical Health Sciences, Doisy College of Health Science, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Aravinda Ganapathy
- Department of Clinical Health Sciences, Doisy College of Health Science, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Kyle Pemberton
- Department of Biology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| | - Eric Tycksen
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, United States
| | - Amelia K Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - James D Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - David B Beck
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Wendy K Chung
- Department of Pediatrics and Medicine, Columbia University Medical Center, New York, NY, United States
| | - Christina A Gurnett
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - G Chinnadurai
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| |
Collapse
|
14
|
Yang G, Bondaruk J, Cogdell D, Wang Z, Lee S, Lee JG, Zhang S, Choi W, Wang Y, Liang Y, Wang G, Wang Y, Yao H, Dadhania V, Gao J, Logothetis C, Siefker-Radtke A, Kamat A, Dinney C, Theodorescu D, Kimmel M, Wei P, Guo CC, Weinstein JN, McConkey DJ, Czerniak B. Urothelial-to-Neural Plasticity Drives Progression to Small Cell Bladder Cancer. iScience 2020; 23:101201. [PMID: 32521509 PMCID: PMC7286965 DOI: 10.1016/j.isci.2020.101201] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/08/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
We report a comprehensive molecular analysis of 34 cases of small cell carcinoma (SCC) and 84 cases of conventional urothelial carcinoma (UC), with The Cancer Genome Atlas cohort of 408 conventional UC bladder cancers used as the reference. SCCs showed mutational landscapes characterized by nearly uniform inactivation of TP53 and were dominated by Sanger mutation signature 3 associated with loss of BRCA1/2 function. SCCs were characterized by downregulation of luminal and basal markers and were referred to as double-negative. Transcriptome analyses indicated that SCCs displayed lineage plasticity driven by a urothelial-to-neural phenotypic switch with a dysregulated epithelial-to-mesenchymal transition network. SCCs were depleted of immune cells, and expressed high levels of the immune checkpoint receptor, adenosine receptor A2A (ADORA2A), which is a potent inhibitor of immune infiltration. Our observations have important implications for the prognostication and development of more effective therapies for this lethal bladder cancer variant. SCCs show TP53/RB1 loss with mutational signature of BRCA1/2 loss of function SCCs are driven by neural phenotypic switch with dysregulated EMT network SCCs show depleted immune phenotype with upregulation of ADORA2A
Collapse
Affiliation(s)
- Guoliang Yang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jolanta Bondaruk
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Cogdell
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ziqiao Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sangkyou Lee
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - June Goo Lee
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shizhen Zhang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Woonyoung Choi
- Johns Hopkins Greenberg Bladder Cancer Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Yan Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yu Liang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gang Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Yao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vipulkumar Dadhania
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arlene Siefker-Radtke
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ashish Kamat
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Colin Dinney
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dan Theodorescu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai, Los Angeles, CA, USA
| | - Marek Kimmel
- Department of Statistics, Rice University, Houston, TX, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Charles C Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - John N Weinstein
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David J McConkey
- Johns Hopkins Greenberg Bladder Cancer Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Bogdan Czerniak
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
Hsu TT, Huang TN, Hsueh YP. Anterior Commissure Regulates Neuronal Activity of Amygdalae and Influences Locomotor Activity, Social Interaction and Fear Memory in Mice. Front Mol Neurosci 2020; 13:47. [PMID: 32296306 PMCID: PMC7136557 DOI: 10.3389/fnmol.2020.00047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
The two hemispheres of the vertebrate brain are connected through several commissures. Although the anterior commissure (AC) is the most conserved white matter structure in the brains of different vertebrates, its complete physiological functionality remains elusive. Since the AC is involved in the connection between two amygdalae and because amygdalae are critical for emotional behaviors and social interaction, we assessed amygdalar activity and function to investigate the physiological role of the AC. We first performed ex vivo electrophysiological recording on mouse brains to demonstrate that the AC delivers a positive signal to facilitate synaptic responses and to recruit basolateral amygdalar neurons via glutamatergic synapses. Transection was then undertaken to investigate the role of the AC in vivo. Results from in vivo optogenetic stimulation suggest that AC transection impairs mutual activation between two basolateral amygdalae. Behavioral analyses were then used to assess if AC surgical lesioning results in hyperactivity, anxiety, social reduction or learning/memory impairment, which are behavioral features associated with neuropsychiatric disorders, such as autism spectrum disorders. We found that AC transection results in higher locomotor activity, aberrant social interaction and reduced associative memory, but not anxiety. Moreover, systemic administration of D-cycloserine, a coagonist of N-methyl-D-aspartate receptor, ameliorated auditory fear memory in AC-transected mice, reinforcing our evidence that the AC potentiates the activity of basolateral amygdalae. Our study suggests that the AC regulates basolateral amygdalar activity and influences neuropsychiatry-related behaviors in mice.
Collapse
Affiliation(s)
- Tsan-Ting Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Tzyy-Nan Huang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
16
|
Greene AG, Eivers SB, Dervan EWJ, O'Brien CJ, Wallace DM. Lysyl Oxidase Like 1: Biological roles and regulation. Exp Eye Res 2020; 193:107975. [PMID: 32070696 DOI: 10.1016/j.exer.2020.107975] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/12/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
Lysyl Oxidase Like 1 (LOXL1) is a gene that encodes for the LOXL1 enzyme. This enzyme is required for elastin biogenesis and collagen cross-linking, polymerising tropoelastin monomers into elastin polymers. Its main role is in elastin homeostasis and matrix remodelling during injury, fibrosis and cancer development. Because of its vast range of biological functions, abnormalities in LOXL1 underlie many disease processes. Decreased LOXL1 expression is observed in disorders of elastin such as Cutis Laxa and increased expression is reported in fibrotic disease such as Idiopathic Pulmonary Fibrosis. LOXL1 is also downregulated in the lamina cribrosa in pseudoexfoliation glaucoma and genetic variants in the LOXL1 gene have been linked with an increased risk of developing pseudoexfoliation glaucoma and pseudoexfoliation syndrome. However the two major risk alleles are reversed in certain ethnic groups and are present in a large proportion of the normal population, implying complex genetic and environmental regulation is involved in disease pathogenesis. It also appears that the non-coding variants in intron 1 of LOXL1 may be involved in the regulation of LOXL1 expression. Gene alteration may occur via a number of epigenetic and post translational mechanisms such as DNA methylation, long non-coding RNAs and microRNAs. These may represent future therapeutic targets for disease. Environmental factors such as hypoxia, oxidative stress and ultraviolet radiation exposure alter LOXL1 expression, and it is likely a combination of these genetic and environmental factors that influence disease development and progression. In this review, we discuss LOXL1 properties, biological roles and regulation in detail with a focus on pseudoexfoliation syndrome and glaucoma.
Collapse
Affiliation(s)
- Alison G Greene
- UCD Clinical Research Centre, School of Medicine, University College Dublin, Ireland.
| | - Sarah B Eivers
- UCD Clinical Research Centre, School of Medicine, University College Dublin, Ireland
| | - Edward W J Dervan
- Dept. of Ophthalmology, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Colm J O'Brien
- UCD Clinical Research Centre, School of Medicine, University College Dublin, Ireland; Dept. of Ophthalmology, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Deborah M Wallace
- UCD Clinical Research Centre, School of Medicine, University College Dublin, Ireland
| |
Collapse
|
17
|
Ward J, Lyall LM, Bethlehem RAI, Ferguson A, Strawbridge RJ, Lyall DM, Cullen B, Graham N, Johnston KJA, Bailey MES, Murray GK, Smith DJ. Novel genome-wide associations for anhedonia, genetic correlation with psychiatric disorders, and polygenic association with brain structure. Transl Psychiatry 2019; 9:327. [PMID: 31797917 PMCID: PMC6892870 DOI: 10.1038/s41398-019-0635-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/18/2019] [Accepted: 10/20/2019] [Indexed: 12/20/2022] Open
Abstract
Anhedonia is a core symptom of several psychiatric disorders but its biological underpinnings are poorly understood. We performed a genome-wide association study of state anhedonia in 375,275 UK Biobank participants and assessed for genetic correlation between anhedonia and neuropsychiatric conditions (major depressive disorder, schizophrenia, bipolar disorder, obsessive compulsive disorder and Parkinson's Disease). We then used a polygenic risk score approach to test for association between genetic loading for anhedonia and both brain structure and brain function. This included: magnetic resonance imaging (MRI) assessments of total grey matter volume, white matter volume, cerebrospinal fluid volume, and 15 cortical/subcortical regions of interest; diffusion tensor imaging (DTI) measures of white matter tract integrity; and functional MRI activity during an emotion processing task. We identified 11 novel loci associated at genome-wide significance with anhedonia, with a SNP heritability estimate (h2SNP) of 5.6%. Strong positive genetic correlations were found between anhedonia and major depressive disorder, schizophrenia and bipolar disorder; but not with obsessive compulsive disorder or Parkinson's Disease. Polygenic risk for anhedonia was associated with poorer brain white matter integrity, smaller total grey matter volume, and smaller volumes of brain regions linked to reward and pleasure processing, including orbito-frontal cortex. In summary, the identification of novel anhedonia-associated loci substantially expands our current understanding of the biological basis of state anhedonia and genetic correlations with several psychiatric disorders confirm the utility of this phenotype as a transdiagnostic marker of vulnerability to mental illness. We also provide the first evidence that genetic risk for state anhedonia influences brain structure, including in regions associated with reward and pleasure processing.
Collapse
Affiliation(s)
- Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Amy Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Rona J Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Breda Cullen
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Nicholas Graham
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Mark E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Graham K Murray
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
| |
Collapse
|
18
|
Schafer ST, Paquola ACM, Stern S, Gosselin D, Ku M, Pena M, Kuret TJM, Liyanage M, Mansour AA, Jaeger BN, Marchetto MC, Glass CK, Mertens J, Gage FH. Pathological priming causes developmental gene network heterochronicity in autistic subject-derived neurons. Nat Neurosci 2019; 22:243-255. [PMID: 30617258 PMCID: PMC6402576 DOI: 10.1038/s41593-018-0295-x] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022]
Abstract
Autism spectrum disorder (ASD) is thought to emerge during early cortical development. However, the exact developmental stages and associated molecular networks that prime disease propensity are elusive. To profile early neurodevelopmental alterations in ASD with macrocephaly, we monitored subject-derived induced pluripotent stem cells (iPSCs) throughout the recapitulation of cortical development. Our analysis revealed ASD-associated changes in the maturational sequence of early neuron development, involving temporal dysregulation of specific gene networks and morphological growth acceleration. The observed changes tracked back to a pathologically primed stage in neural stem cells (NSCs), reflected by altered chromatin accessibility. Concerted over-representation of network factors in control NSCs was sufficient to trigger ASD-like features, and circumventing the NSC stage by direct conversion of ASD iPSCs into induced neurons abolished ASD-associated phenotypes. Our findings identify heterochronic dynamics of a gene network that, while established earlier in development, contributes to subsequent neurodevelopmental aberrations in ASD.
Collapse
Affiliation(s)
- Simon T Schafer
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Apua C M Paquola
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shani Stern
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - David Gosselin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, Canada
| | - Manching Ku
- Next Generation Sequencing Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Monique Pena
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Thomas J M Kuret
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Marvin Liyanage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Abed AlFatah Mansour
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Baptiste N Jaeger
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Maria C Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jerome Mertens
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Department of Genomics, Stem Cell Biology and Regenerative Medicine, Institute of Molecular Biology & CMBI, University of Innsbruck, Innsbruck, Austria
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
19
|
A novel ISLR2-linked autosomal recessive syndrome of congenital hydrocephalus, arthrogryposis and abdominal distension. Hum Genet 2018; 138:105-107. [DOI: 10.1007/s00439-018-1963-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 11/21/2018] [Indexed: 01/22/2023]
|
20
|
Martin-Lopez E, Meller SJ, Greer CA. Development of piriform cortex interhemispheric connections via the anterior commissure: progressive and regressive strategies. Brain Struct Funct 2018; 223:4067-4085. [PMID: 30141078 DOI: 10.1007/s00429-018-1741-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/21/2018] [Indexed: 12/27/2022]
Abstract
The anterior commissure (AC) is a phylogenetically conserved inter-hemispheric connection found among vertebrates with bilateral symmetry. The AC connects predominantly olfactory areas but many aspects of its development and structure are unknown. To fill this gap, we investigated the embryonic and postnatal development of the AC by tracing axons with DiI and the piggyback transposon multicolor system. With this strategy, we show that axon growth during establishment of the AC follows a strictly regulated timeline of events that include waiting periods ("regressive strategies") as well as periods of active axon outgrowth ("progressive strategies"). We also provide evidence that these processes may be regulated in the midline via overexpression of chondroitin sulfate proteoglycans. Additionally, we demonstrate that the ipsi- and contralateral innervation of piriform cortex occurs simultaneously. Morphologically, we found that 20% of axons were myelinated by postnatal day (P) 22, in a process that occurred fundamentally around P14. By immunohistochemistry, we described the presence of glial cells and two new subtypes of neurons: one expressing a calretinin (CR)-/MAP2+ phenotype, distributed homogeneously inside the AC; and the other expressing a CR+/MAP2+ phenotype that lies beneath the bed nucleus of the stria terminalis. Our results are consistent with the notion that the AC follows a strictly regulated program during the embryonic and postnatal development similarly to other distal targeting axonal tracts.
Collapse
Affiliation(s)
- Eduardo Martin-Lopez
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.,Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Sarah J Meller
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.,Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Charles A Greer
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA. .,Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA. .,The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|