1
|
Zaragoza MV, Bui TA, Widyastuti HP, Mehrabi M, Cang Z, Sha Y, Grosberg A, Nie Q. LMNA-Related Dilated Cardiomyopathy: Single-Cell Transcriptomics during Patient-Derived iPSC Differentiation Support Cell Type and Lineage-Specific Dysregulation of Gene Expression and Development for Cardiomyocytes and Epicardium-Derived Cells with Lamin A/C Haploinsufficiency. Cells 2024; 13:1479. [PMID: 39273049 PMCID: PMC11394257 DOI: 10.3390/cells13171479] [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: 06/12/2024] [Revised: 08/14/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
LMNA-related dilated cardiomyopathy (DCM) is an autosomal-dominant genetic condition with cardiomyocyte and conduction system dysfunction often resulting in heart failure or sudden death. The condition is caused by mutation in the Lamin A/C (LMNA) gene encoding Type-A nuclear lamin proteins involved in nuclear integrity, epigenetic regulation of gene expression, and differentiation. The molecular mechanisms of the disease are not completely understood, and there are no definitive treatments to reverse progression or prevent mortality. We investigated possible mechanisms of LMNA-related DCM using induced pluripotent stem cells derived from a family with a heterozygous LMNA c.357-2A>G splice-site mutation. We differentiated one LMNA-mutant iPSC line derived from an affected female (Patient) and two non-mutant iPSC lines derived from her unaffected sister (Control) and conducted single-cell RNA sequencing for 12 samples (four from Patients and eight from Controls) across seven time points: Day 0, 2, 4, 9, 16, 19, and 30. Our bioinformatics workflow identified 125,554 cells in raw data and 110,521 (88%) high-quality cells in sequentially processed data. Unsupervised clustering, cell annotation, and trajectory inference found complex heterogeneity: ten main cell types; many possible subtypes; and lineage bifurcation for cardiac progenitors to cardiomyocytes (CMs) and epicardium-derived cells (EPDCs). Data integration and comparative analyses of Patient and Control cells found cell type and lineage-specific differentially expressed genes (DEGs) with enrichment, supporting pathway dysregulation. Top DEGs and enriched pathways included 10 ZNF genes and RNA polymerase II transcription in pluripotent cells (PP); BMP4 and TGF Beta/BMP signaling, sarcomere gene subsets and cardiogenesis, CDH2 and EMT in CMs; LMNA and epigenetic regulation, as well as DDIT4 and mTORC1 signaling in EPDCs. Top DEGs also included XIST and other X-linked genes, six imprinted genes (SNRPN, PWAR6, NDN, PEG10, MEG3, MEG8), and enriched gene sets related to metabolism, proliferation, and homeostasis. We confirmed Lamin A/C haploinsufficiency by allelic expression and Western blot. Our complex Patient-derived iPSC model for Lamin A/C haploinsufficiency in PP, CM, and EPDC provided support for dysregulation of genes and pathways, many previously associated with Lamin A/C defects, such as epigenetic gene expression, signaling, and differentiation. Our findings support disruption of epigenomic developmental programs, as proposed in other LMNA disease models. We recognized other factors influencing epigenetics and differentiation; thus, our approach needs improvement to further investigate this mechanism in an iPSC-derived model.
Collapse
Affiliation(s)
- Michael V. Zaragoza
- UCI Cardiogenomics Program, Pediatrics and Biological Chemistry, UC Irvine School of Medicine, Irvine, CA 92697, USA
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Thuy-Anh Bui
- UCI Cardiogenomics Program, Pediatrics and Biological Chemistry, UC Irvine School of Medicine, Irvine, CA 92697, USA
| | - Halida P. Widyastuti
- UCI Cardiogenomics Program, Pediatrics and Biological Chemistry, UC Irvine School of Medicine, Irvine, CA 92697, USA
| | - Mehrsa Mehrabi
- Biomedical Engineering and Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Zixuan Cang
- Mathematics and NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Yutong Sha
- Mathematics and NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Anna Grosberg
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
- Biomedical Engineering and Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Qing Nie
- Mathematics and NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
2
|
Zaragoza MV, Bui TA, Widyastuti HP, Mehrabi M, Cang Z, Sha Y, Grosberg A, Nie Q. LMNA -Related Dilated Cardiomyopathy: Single-Cell Transcriptomics during Patient-derived iPSC Differentiation Support Cell type and Lineage-specific Dysregulation of Gene Expression and Development for Cardiomyocytes and Epicardium-Derived Cells with Lamin A/C Haploinsufficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598335. [PMID: 38915555 PMCID: PMC11195187 DOI: 10.1101/2024.06.12.598335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
LMNA -Related Dilated Cardiomyopathy (DCM) is an autosomal-dominant genetic condition with cardiomyocyte and conduction system dysfunction often resulting in heart failure or sudden death. The condition is caused by mutation in the Lamin A/C ( LMNA ) gene encoding Type-A nuclear lamin proteins involved in nuclear integrity, epigenetic regulation of gene expression, and differentiation. Molecular mechanisms of disease are not completely understood, and there are no definitive treatments to reverse progression or prevent mortality. We investigated possible mechanisms of LMNA -Related DCM using induced pluripotent stem cells derived from a family with a heterozygous LMNA c.357-2A>G splice-site mutation. We differentiated one LMNA mutant iPSC line derived from an affected female (Patient) and two non-mutant iPSC lines derived from her unaffected sister (Control) and conducted single-cell RNA sequencing for 12 samples (4 Patient and 8 Control) across seven time points: Day 0, 2, 4, 9, 16, 19, and 30. Our bioinformatics workflow identified 125,554 cells in raw data and 110,521 (88%) high-quality cells in sequentially processed data. Unsupervised clustering, cell annotation, and trajectory inference found complex heterogeneity: ten main cell types; many possible subtypes; and lineage bifurcation for Cardiac Progenitors to Cardiomyocytes (CM) and Epicardium-Derived Cells (EPDC). Data integration and comparative analyses of Patient and Control cells found cell type and lineage differentially expressed genes (DEG) with enrichment to support pathway dysregulation. Top DEG and enriched pathways included: 10 ZNF genes and RNA polymerase II transcription in Pluripotent cells (PP); BMP4 and TGF Beta/BMP signaling, sarcomere gene subsets and cardiogenesis, CDH2 and EMT in CM; LMNA and epigenetic regulation and DDIT4 and mTORC1 signaling in EPDC. Top DEG also included: XIST and other X-linked genes, six imprinted genes: SNRPN , PWAR6 , NDN , PEG10 , MEG3 , MEG8 , and enriched gene sets in metabolism, proliferation, and homeostasis. We confirmed Lamin A/C haploinsufficiency by allelic expression and Western blot. Our complex Patient-derived iPSC model for Lamin A/C haploinsufficiency in PP, CM, and EPDC provided support for dysregulation of genes and pathways, many previously associated with Lamin A/C defects, such as epigenetic gene expression, signaling, and differentiation. Our findings support disruption of epigenomic developmental programs as proposed in other LMNA disease models. We recognized other factors influencing epigenetics and differentiation; thus, our approach needs improvement to further investigate this mechanism in an iPSC-derived model.
Collapse
|
3
|
McCoy MD, Sarasua SM, DeLuca JM, Davis S, Rogers RC, Phelan K, Boccuto L. Genetics of kidney disorders in Phelan-McDermid syndrome: evidence from 357 registry participants. Pediatr Nephrol 2024; 39:749-760. [PMID: 37733098 DOI: 10.1007/s00467-023-06146-y] [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: 04/13/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS) is a rare genetic disorder caused by SHANK3 pathogenic variants or chromosomal rearrangements affecting the chromosome 22q13 region. Previous research found that kidney disorders, primarily congenital anomalies of the kidney and urinary tract, are common in people with PMS, yet research into candidate genes has been hampered by small study sizes and lack of attention to these problems. METHODS We used a cohort of 357 people from the Phelan-McDermid Syndrome Foundation International Registry to investigate the prevalence of kidney disorders in PMS using a cross-sectional design and to identify 22q13 genes contributing to these disorders. RESULTS Kidney disorders reported included vesicoureteral reflux (n = 37), hydronephrosis (n = 36), dysplastic kidneys (n = 19), increased kidney size (n = 19), polycystic kidneys (15 cases), and kidney stones (n = 4). Out of 315 subjects with a 22q13 deletion, 101 (32%) had at least one kidney disorder, while only one out of 42 (2%) individuals with a SHANK3 pathogenic variant had a kidney disorder (increased kidney size). We identified two genomic regions that were significantly associated with having a kidney disorder with the peak associations observed near positions approximately 5 Mb and 400 Kb from the telomere. CONCLUSIONS The candidate genes for kidney disorders include FBLN1, WNT7B, UPK3A, CELSR1, and PLXNB2. This study demonstrates the utility of patient registries for uncovering genetic contributions to rare diseases. Future work should focus on functional studies for these genes to assess their potential pathogenic contribution to the different subsets of kidney disorders.
Collapse
Affiliation(s)
- Megan D McCoy
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA
| | - Sara M Sarasua
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA.
| | - Jane M DeLuca
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA
| | - Stephanie Davis
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA
| | | | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists and Research Institute, Fort Myers, FL, 33916, USA
| | - Luigi Boccuto
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA
| |
Collapse
|
4
|
Cismaru CA, Tomuleasa C, Jurj A, Chira S, Isachekcu E, Cismaru G, Gherman LM, Gulei D, Munteanu R, Berindan Neagoe I. Synergistic Effect of Human Chorionic Gonadotropin and Granulocyte Colony Stimulating Factor in the Mobilization of HSPCs Improves Overall Survival After PBSCT in a Preclinical Murine Model. Are We Far Enough for Therapy? Stem Cell Rev Rep 2024; 20:206-217. [PMID: 37922107 DOI: 10.1007/s12015-023-10648-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 11/05/2023]
Abstract
Strategies to improve hematopoietic stem and progenitor cell (HSPC) mobilization from the bone marrow can have a pivotal role in addressing iatrogenic bone-marrow insufficiency from chemo(radio)therapy and overcoming peripheral blood stem cell transplantation (PBSCT) limitations such as insufficient mobilization. Granulocyte-colony stimulating factor (G-CSF) represents the standard mobilization strategy for HSPC and has done so for more than three decades since its FDA approval. Its association with non-G-CSF agents is often employed for difficult HSPC mobilization. However, obtaining a synergistic effect between the two classes is limited by different timing and mechanisms of action. Based on our previous in vitro results, we tested the mobilization potential of human chorionic gonadotropin (HCG), alone and in combination with G-CSF in vivo in a murine study. Our results show an improved mobilization capability of the combination, which seems to act synergistically in stimulating hematopoiesis. With the current understanding of the dynamics of HSPCs and their origins in more primitive cells related to the germline, new strategies to employ the mobilization of hematopoietic progenitors using chorionic gonadotropins could soon become clinical practice.
Collapse
Affiliation(s)
- Cosmin Andrei Cismaru
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, P.O. 400393, Cluj-Napoca, Romania.
| | - Ciprian Tomuleasa
- MEDFUTURE - The Research Center for Advanced Medicine "Iuliu Hatieganu", University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, P.O. 400393, Cluj-Napoca, Romania
| | - Sergiu Chira
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, P.O. 400393, Cluj-Napoca, Romania
| | - Ekaterina Isachekcu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, P.O. 400393, Cluj-Napoca, Romania
| | - Gabriel Cismaru
- Department of Internal Medicine, Cardiology-Rehabilitation, "Iuliu Hatieganu", University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Luciana Madalina Gherman
- Laboratory Animal Facility - Centre for Experimental Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- MEDFUTURE - The Research Center for Advanced Medicine "Iuliu Hatieganu", University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Raluca Munteanu
- MEDFUTURE - The Research Center for Advanced Medicine "Iuliu Hatieganu", University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, P.O. 400393, Cluj-Napoca, Romania
| |
Collapse
|
5
|
Licata L, Via A, Turina P, Babbi G, Benevenuta S, Carta C, Casadio R, Cicconardi A, Facchiano A, Fariselli P, Giordano D, Isidori F, Marabotti A, Martelli PL, Pascarella S, Pinelli M, Pippucci T, Russo R, Savojardo C, Scafuri B, Valeriani L, Capriotti E. Resources and tools for rare disease variant interpretation. Front Mol Biosci 2023; 10:1169109. [PMID: 37234922 PMCID: PMC10206239 DOI: 10.3389/fmolb.2023.1169109] [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: 02/18/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Collectively, rare genetic disorders affect a substantial portion of the world's population. In most cases, those affected face difficulties in receiving a clinical diagnosis and genetic characterization. The understanding of the molecular mechanisms of these diseases and the development of therapeutic treatments for patients are also challenging. However, the application of recent advancements in genome sequencing/analysis technologies and computer-aided tools for predicting phenotype-genotype associations can bring significant benefits to this field. In this review, we highlight the most relevant online resources and computational tools for genome interpretation that can enhance the diagnosis, clinical management, and development of treatments for rare disorders. Our focus is on resources for interpreting single nucleotide variants. Additionally, we present use cases for interpreting genetic variants in clinical settings and review the limitations of these results and prediction tools. Finally, we have compiled a curated set of core resources and tools for analyzing rare disease genomes. Such resources and tools can be utilized to develop standardized protocols that will enhance the accuracy and effectiveness of rare disease diagnosis.
Collapse
Affiliation(s)
- Luana Licata
- Department of Biology, University of Rome Tor Vergata, Roma, Italy
| | - Allegra Via
- Department of Biochemical Sciences “A. Rossi Fanelli”, University of Rome “La Sapienza”, Roma, Italy
| | - Paola Turina
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giulia Babbi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Claudio Carta
- National Centre for Rare Diseases, Istituto Superiore di Sanità, Roma, Italy
| | - Rita Casadio
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Andrea Cicconardi
- Department of Physics, University of Genova, Genova, Italy
- Italiano di Tecnologia—IIT, Genova, Italy
| | - Angelo Facchiano
- National Research Council, Institute of Food Science, Avellino, Italy
| | - Piero Fariselli
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Deborah Giordano
- National Research Council, Institute of Food Science, Avellino, Italy
| | - Federica Isidori
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Anna Marabotti
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Fisciano, SA, Italy
| | - Pier Luigi Martelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Stefano Pascarella
- Department of Biochemical Sciences “A. Rossi Fanelli”, University of Rome “La Sapienza”, Roma, Italy
| | - Michele Pinelli
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Napoli, Italy
| | - Tommaso Pippucci
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Roberta Russo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, Napoli, Italy
| | - Castrense Savojardo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Bernardina Scafuri
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Fisciano, SA, Italy
| | | | - Emidio Capriotti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
6
|
Rockweiler NB, Ramu A, Nagirnaja L, Wong WH, Noordam MJ, Drubin CW, Huang N, Miller B, Todres EZ, Vigh-Conrad KA, Zito A, Small KS, Ardlie KG, Cohen BA, Conrad DF. The origins and functional effects of postzygotic mutations throughout the human life span. Science 2023; 380:eabn7113. [PMID: 37053313 PMCID: PMC11246725 DOI: 10.1126/science.abn7113] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/17/2023] [Indexed: 04/15/2023]
Abstract
Postzygotic mutations (PZMs) begin to accrue in the human genome immediately after fertilization, but how and when PZMs affect development and lifetime health remain unclear. To study the origins and functional consequences of PZMs, we generated a multitissue atlas of PZMs spanning 54 tissue and cell types from 948 donors. Nearly half the variation in mutation burden among tissue samples can be explained by measured technical and biological effects, and 9% can be attributed to donor-specific effects. Through phylogenetic reconstruction of PZMs, we found that their type and predicted functional impact vary during prenatal development, across tissues, and through the germ cell life cycle. Thus, methods for interpreting effects across the body and the life span are needed to fully understand the consequences of genetic variants.
Collapse
Affiliation(s)
- Nicole B. Rockweiler
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Present address: Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Avinash Ramu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Liina Nagirnaja
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Wing H. Wong
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Present Address: Departments of Genetics and Medicine, Stanford University, CA 94305, USA
| | - Michiel J. Noordam
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Casey W. Drubin
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ni Huang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Present Address: T-Therapeutics Ltd., Cambridge CB21 6AD, UK
| | - Brian Miller
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Ellen Z. Todres
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Katinka A. Vigh-Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Antonino Zito
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
- Present Address: Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114, USA; Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Kerrin S. Small
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | | | - Barak A. Cohen
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Donald F. Conrad
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
- Center for Embryonic Cell & Gene Therapy, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
7
|
Alsaedi SB, Mineta K, Gao X, Gojobori T. Computational network analysis of host genetic risk variants of severe COVID-19. Hum Genomics 2023; 17:17. [PMID: 36859360 PMCID: PMC9977643 DOI: 10.1186/s40246-023-00454-y] [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: 11/24/2022] [Accepted: 01/28/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Genome-wide association studies have identified numerous human host genetic risk variants that play a substantial role in the host immune response to SARS-CoV-2. Although these genetic risk variants significantly increase the severity of COVID-19, their influence on body systems is poorly understood. Therefore, we aim to interpret the biological mechanisms and pathways associated with the genetic risk factors and immune responses in severe COVID-19. We perform a deep analysis of previously identified risk variants and infer the hidden interactions between their molecular networks through disease mapping and the similarity of the molecular functions between constructed networks. RESULTS We designed a four-stage computational workflow for systematic genetic analysis of the risk variants. We integrated the molecular profiles of the risk factors with associated diseases, then constructed protein-protein interaction networks. We identified 24 protein-protein interaction networks with 939 interactions derived from 109 filtered risk variants in 60 risk genes and 56 proteins. The majority of molecular functions, interactions and pathways are involved in immune responses; several interactions and pathways are related to the metabolic and cardiovascular systems, which could lead to multi-organ complications and dysfunction. CONCLUSIONS This study highlights the importance of analyzing molecular interactions and pathways to understand the heterogeneous susceptibility of the host immune response to SARS-CoV-2. We propose new insights into pathogenicity analysis of infections by including genetic risk information as essential factors to predict future complications during and after infection. This approach may assist more precise clinical decisions and accurate treatment plans to reduce COVID-19 complications.
Collapse
Affiliation(s)
- Sakhaa B. Alsaedi
- grid.45672.320000 0001 1926 5090Division of Computer, Electrical and Mathematical Sciences and Engineering, Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia ,grid.412892.40000 0004 1754 9358College of Computer Science and Engineering (CCSE), Taibah University, Medina, Saudi Arabia
| | - Katsuhiko Mineta
- grid.45672.320000 0001 1926 5090Division of Computer, Electrical and Mathematical Sciences and Engineering, Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia ,grid.5290.e0000 0004 1936 9975AND Research Organization for Nano and Life Innovation, Waseda University, Tokyo, 162-0041 Japan
| | - Xin Gao
- grid.45672.320000 0001 1926 5090Division of Computer, Electrical and Mathematical Sciences and Engineering, Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia
| | - Takashi Gojobori
- Division of Computer, Electrical and Mathematical Sciences and Engineering, Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
8
|
Vrij EJ, Scholte op Reimer YS, Fuentes LR, Guerreiro IM, Holzmann V, Aldeguer JF, Sestini G, Koo BK, Kind J, van Blitterswijk CA, Rivron NC. A pendulum of induction between the epiblast and extra-embryonic endoderm supports post-implantation progression. Development 2022; 149:dev192310. [PMID: 35993866 PMCID: PMC9534490 DOI: 10.1242/dev.192310] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/23/2022] [Indexed: 08/17/2023]
Abstract
Embryogenesis is supported by dynamic loops of cellular interactions. Here, we create a partial mouse embryo model to elucidate the principles of epiblast (Epi) and extra-embryonic endoderm co-development (XEn). We trigger naive mouse embryonic stem cells to form a blastocyst-stage niche of Epi-like cells and XEn-like cells (3D, hydrogel free and serum free). Once established, these two lineages autonomously progress in minimal medium to form an inner pro-amniotic-like cavity surrounded by polarized Epi-like cells covered with visceral endoderm (VE)-like cells. The progression occurs through reciprocal inductions by which the Epi supports the primitive endoderm (PrE) to produce a basal lamina that subsequently regulates Epi polarization and/or cavitation, which, in return, channels the transcriptomic progression to VE. This VE then contributes to Epi bifurcation into anterior- and posterior-like states. Similarly, boosting the formation of PrE-like cells within blastoids supports developmental progression. We argue that self-organization can arise from lineage bifurcation followed by a pendulum of induction that propagates over time.
Collapse
Affiliation(s)
- Erik J. Vrij
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, Netherlands
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Yvonne S. Scholte op Reimer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Laury Roa Fuentes
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, Netherlands
| | - Isabel Misteli Guerreiro
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, UtrechtUppsalalaan 8, 3584 CT Utrecht, Netherlands
| | - Viktoria Holzmann
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Javier Frias Aldeguer
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, Netherlands
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, UtrechtUppsalalaan 8, 3584 CT Utrecht, Netherlands
| | - Giovanni Sestini
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Jop Kind
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, UtrechtUppsalalaan 8, 3584 CT Utrecht, Netherlands
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, Netherlands
| | - Clemens A. van Blitterswijk
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, Netherlands
| | - Nicolas C. Rivron
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| |
Collapse
|
9
|
Association between cancer genes and germ layer specificity. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:218. [PMID: 36175592 DOI: 10.1007/s12032-022-01823-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/14/2022] [Indexed: 10/14/2022]
Abstract
Cancer signaling pathways defining cell fates are related to differentiation. During the developmental process, three germ layers (endoderm, mesoderm, and ectoderm) are formed during embryonic development that differentiate into organs via the epigenetic regulation of specific genes. To examine the relationship, the specificities of cancer gene mutations that depend on the germ layers are studied. The major organs affected by cancer were determined based on statistics from the National Cancer Information Center of Korea, and were grouped according to their germ layer origins. Then, the gene mutation frequencies were evaluated to identify any bias based on the differentiation group using the Catalogue of Somatic Mutations in Cancer (COSMIC) database. The chi-square test showed that the p-value of 152 of 166 genes was less than 0.05, and 151 genes showed p-values of less than 0.05 even after adjusting for the false discovery rate (FDR). The germ layer-specific genes were evaluated using visualization based on basic statistics, and the results matched the top ranking genes depending on organs in the COSMIC database.The current study confirmed the germ layer specificity of major cancer genes. The germ layer specificity of mutated driver genes is possibly important in cancer treatments because each mutated gene may react differently depending on the germ layer of origin. By understanding the mechanism of gene mutation in the development and progression of cancer in the context of cell-fate pathways, a more effective therapeutic strategy for cancer can be established.
Collapse
|
10
|
Martin PCN, Kim H, Lövkvist C, Hong B, Won KJ. Vesalius: high-resolution in silico anatomization of spatial transcriptomic data using image analysis. Mol Syst Biol 2022; 18:e11080. [PMID: 36065846 PMCID: PMC9446088 DOI: 10.15252/msb.202211080] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/25/2022] Open
Abstract
Characterization of tissue architecture promises to deliver insights into development, cell communication, and disease. In silico spatial domain retrieval methods have been developed for spatial transcriptomics (ST) data assuming transcriptional similarity of neighboring barcodes. However, domain retrieval approaches with this assumption cannot work in complex tissues composed of multiple cell types. This task becomes especially challenging in cellular resolution ST methods. We developed Vesalius to decipher tissue anatomy from ST data by applying image processing technology. Vesalius uniquely detected territories composed of multiple cell types and successfully recovered tissue structures in high-resolution ST data including in mouse brain, embryo, liver, and colon. Utilizing this tissue architecture, Vesalius identified tissue morphology-specific gene expression and regional specific gene expression changes for astrocytes, interneuron, oligodendrocytes, and entorhinal cells in the mouse brain.
Collapse
Affiliation(s)
- Patrick C N Martin
- Department of Computational BiomedicineCedars‐Sinai Medical CenterHollywoodCAUSA
- Biotech Research and Innovation Centre (BRIC)University of CopenhagenCopenhagenDenmark
| | - Hyobin Kim
- Department of Computational BiomedicineCedars‐Sinai Medical CenterHollywoodCAUSA
- Biotech Research and Innovation Centre (BRIC)University of CopenhagenCopenhagenDenmark
| | - Cecilia Lövkvist
- Biotech Research and Innovation Centre (BRIC)University of CopenhagenCopenhagenDenmark
| | - Byung‐Woo Hong
- Computer Science DepartmentChung‐Ang UniversitySeoulKorea
| | - Kyoung Jae Won
- Department of Computational BiomedicineCedars‐Sinai Medical CenterHollywoodCAUSA
- Biotech Research and Innovation Centre (BRIC)University of CopenhagenCopenhagenDenmark
| |
Collapse
|
11
|
Braun M, Shoshani S, Tabach Y. Transcriptome changes in DM1 patients’ tissues are governed by the RNA interference pathway. Front Mol Biosci 2022; 9:955753. [PMID: 36060259 PMCID: PMC9437208 DOI: 10.3389/fmolb.2022.955753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic disease caused by pathogenic expansions of CTG repeats. The expanded repeats are transcribed to long RNA and induce cellular toxicity. Recent studies suggest that the CUG repeats are processed by the RNA interference (RNAi) pathway to generate small interfering repeated RNA (siRNA). However, the effects of the CTG repeat-derived siRNAs remain unclear. We hypothesize that the RNAi machinery in DM1 patients generates distinct gene expression patterns that determine the disease phenotype in the individual patient. The abundance of genes with complementary repeats that are targeted by siRNAs in each tissue determines the way that the tissue is affected in DM1. We integrated and analyzed published transcriptome data from muscle, heart, and brain biopsies of DM1 patients, and revealed shared, characteristic changes that correlated with disease phenotype. These signatures are overrepresented by genes and transcription factors bearing endogenous CTG/CAG repeats and are governed by aberrant activity of the RNAi machinery, miRNAs, and a specific gain-of-function of the CTG repeats. Computational analysis of the DM1 transcriptome enhances our understanding of the complex pathophysiology of the disease and may reveal a path for cure.
Collapse
|
12
|
Generation and characterization of stable pig pregastrulation epiblast stem cell lines. Cell Res 2022; 32:383-400. [PMID: 34848870 PMCID: PMC8976023 DOI: 10.1038/s41422-021-00592-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Pig epiblast-derived pluripotent stem cells are considered to have great potential and broad prospects for human therapeutic model development and livestock breeding. Despite ongoing attempts since the 1990s, no stably defined pig epiblast-derived stem cell line has been established. Here, guided by insights from a large-scale single-cell transcriptome analysis of pig embryos from embryonic day (E) 0 to E14, specifically, the tracing of pluripotency changes during epiblast development, we developed an in vitro culture medium for establishing and maintaining stable pluripotent stem cell lines from pig E10 pregastrulation epiblasts (pgEpiSCs). Enabled by chemical inhibition of WNT-related signaling in combination with growth factors in the FGF/ERK, JAK/STAT3, and Activin/Nodal pathways, pgEpiSCs maintain their pluripotency transcriptome features, similar to those of E10 epiblast cells, and normal karyotypes after more than 240 passages and have the potential to differentiate into three germ layers. Strikingly, ultradeep in situ Hi-C analysis revealed functional impacts of chromatin 3D-spatial associations on the transcriptional regulation of pluripotency marker genes in pgEpiSCs. In practice, we confirmed that pgEpiSCs readily tolerate at least three rounds of successive gene editing and generated cloned gene-edited live piglets. Our findings deliver on the long-anticipated promise of pig pluripotent stem cells and open new avenues for biological research, animal husbandry, and regenerative biomedicine.
Collapse
|
13
|
Jansen C, Paraiso KD, Zhou JJ, Blitz IL, Fish MB, Charney RM, Cho JS, Yasuoka Y, Sudou N, Bright AR, Wlizla M, Veenstra GJC, Taira M, Zorn AM, Mortazavi A, Cho KWY. Uncovering the mesendoderm gene regulatory network through multi-omic data integration. Cell Rep 2022; 38:110364. [PMID: 35172134 PMCID: PMC8917868 DOI: 10.1016/j.celrep.2022.110364] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 10/30/2021] [Accepted: 01/19/2022] [Indexed: 01/01/2023] Open
Abstract
Mesendodermal specification is one of the earliest events in embryogenesis, where cells first acquire distinct identities. Cell differentiation is a highly regulated process that involves the function of numerous transcription factors (TFs) and signaling molecules, which can be described with gene regulatory networks (GRNs). Cell differentiation GRNs are difficult to build because existing mechanistic methods are low throughput, and high-throughput methods tend to be non-mechanistic. Additionally, integrating highly dimensional data composed of more than two data types is challenging. Here, we use linked self-organizing maps to combine chromatin immunoprecipitation sequencing (ChIP-seq)/ATAC-seq with temporal, spatial, and perturbation RNA sequencing (RNA-seq) data from Xenopus tropicalis mesendoderm development to build a high-resolution genome scale mechanistic GRN. We recover both known and previously unsuspected TF-DNA/TF-TF interactions validated through reporter assays. Our analysis provides insights into transcriptional regulation of early cell fate decisions and provides a general approach to building GRNs using highly dimensional multi-omic datasets.
Collapse
Affiliation(s)
- Camden Jansen
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Kitt D Paraiso
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Jeff J Zhou
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Ira L Blitz
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Margaret B Fish
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Rebekah M Charney
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Jin Sun Cho
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Yuuri Yasuoka
- Laboratory for Comprehensive Genomic Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Norihiro Sudou
- Department of Anatomy, School of Medicine, Toho University, Tokyo, Japan
| | - Ann Rose Bright
- Department of Molecular Developmental Biology, Radboud University, Nijmegen, the Netherlands
| | - Marcin Wlizla
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Gert Jan C Veenstra
- Department of Molecular Developmental Biology, Radboud University, Nijmegen, the Netherlands
| | - Masanori Taira
- Department of Biological Sciences, Chuo University, Tokyo, Japan
| | - Aaron M Zorn
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, CA, USA.
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, CA, USA.
| |
Collapse
|
14
|
Long H, Reeves R, Simon MM. Mouse genomic and cellular annotations. Mamm Genome 2022; 33:19-30. [PMID: 35124726 PMCID: PMC8913471 DOI: 10.1007/s00335-021-09936-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/22/2021] [Indexed: 11/28/2022]
Abstract
AbstractMice have emerged as one of the most popular and valuable model organisms in the research of human biology. This is due to their genetic and physiological similarity to humans, short generation times, availability of genetically homologous inbred strains, and relatively easy laboratory maintenance. Therefore, following the release of the initial human reference genome, the generation of the mouse reference genome was prioritised and represented an important scientific resource for the mouse genetics community. In 2002, the Mouse Genome Sequencing Consortium published an initial draft of the mouse reference genome which contained ~ 96% of the euchromatic genome of female C57BL/6 J mice. Almost two decades on from the publication of the initial draft, sequencing efforts have continued to increase the completeness and accuracy of the C57BL/6 J reference genome alongside advances in genome annotation. Additionally new sequencing technologies have provided a wealth of data that has added to the repertoire of annotations associated with traditional genomic annotations. Including but not limited to advances in regulatory elements, the 3D genome and individual cellular states. In this review we focus on the reference genome C57BL/6 J and summarise the different aspects of genomic and cellular annotations, as well as their relevance to mouse genetic research. We denote a genomic annotation as a functional unit of the genome. Cellular annotations are annotations of cell type or state, defined by the transcriptomic expression profile of a cell. Due to the wide-ranging number and diversity of annotations describing the mouse genome, we focus on gene, repeat and regulatory element annotation as well as two relatively new technologies; 3D genome architecture and single-cell sequencing outlining their utility in genetic research and their current challenges.
Collapse
Affiliation(s)
- Helen Long
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, OX11 0RD, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Richard Reeves
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Michelle M Simon
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, OX11 0RD, UK.
| |
Collapse
|
15
|
Gabriel E, Albanna W, Pasquini G, Ramani A, Josipovic N, Mariappan A, Schinzel F, Karch CM, Bao G, Gottardo M, Suren AA, Hescheler J, Nagel-Wolfrum K, Persico V, Rizzoli SO, Altmüller J, Riparbelli MG, Callaini G, Goureau O, Papantonis A, Busskamp V, Schneider T, Gopalakrishnan J. Human brain organoids assemble functionally integrated bilateral optic vesicles. Cell Stem Cell 2021; 28:1740-1757.e8. [PMID: 34407456 DOI: 10.1016/j.stem.2021.07.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/23/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
During embryogenesis, optic vesicles develop from the diencephalon via a multistep process of organogenesis. Using induced pluripotent stem cell (iPSC)-derived human brain organoids, we attempted to simplify the complexities and demonstrate formation of forebrain-associated bilateral optic vesicles, cellular diversity, and functionality. Around day 30, brain organoids attempt to assemble optic vesicles, which develop progressively as visible structures within 60 days. These optic vesicle-containing brain organoids (OVB-organoids) constitute a developing optic vesicle's cellular components, including primitive corneal epithelial and lens-like cells, retinal pigment epithelia, retinal progenitor cells, axon-like projections, and electrically active neuronal networks. OVB-organoids also display synapsin-1, CTIP-positive myelinated cortical neurons, and microglia. Interestingly, various light intensities could trigger photosensitive activity of OVB-organoids, and light sensitivities could be reset after transient photobleaching. Thus, brain organoids have the intrinsic ability to self-organize forebrain-associated primitive sensory structures in a topographically restricted manner and can allow interorgan interaction studies within a single organoid.
Collapse
Affiliation(s)
- Elke Gabriel
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Walid Albanna
- Institute for Neurophysiology, University of Cologne, 50931 Cologne, Germany; Department of Neurosurgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Giovanni Pasquini
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Anand Ramani
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Natasa Josipovic
- Institute of Pathology, University Medicine Göttingen, Georg-August University Göttingen, 37075 Göttingen, Germany; Center for molecular medicine, Cologne, Universität zu Köln, 50931 Köln, Germany
| | - Aruljothi Mariappan
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Friedrich Schinzel
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63116, USA
| | - Guobin Bao
- Institute of Neurophysiology and Cellular Biophysics, University Medicine Göttingen, Georg-August-Universität Göttingen, 37073 Göttingen, Germany
| | - Marco Gottardo
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Ata Alp Suren
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Kerstin Nagel-Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University, 55099 Mainz, Germany
| | - Veronica Persico
- Department of Life Sciences and Medical Biotechnology University of Siena, Siena 53100, Italy
| | - Silvio O Rizzoli
- Institute of Neurophysiology and Cellular Biophysics, University Medicine Göttingen, Georg-August-Universität Göttingen, 37073 Göttingen, Germany
| | - Janine Altmüller
- Cologne Center for Genomics (CCG), Universität zu Köln, Köln, Germany; Center for molecular medicine, Cologne, Universität zu Köln, 50931 Köln, Germany
| | | | - Giuliano Callaini
- Department of Life Sciences and Medical Biotechnology University of Siena, Siena 53100, Italy
| | - Olivier Goureau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Argyris Papantonis
- Institute of Pathology, University Medicine Göttingen, Georg-August University Göttingen, 37075 Göttingen, Germany
| | - Volker Busskamp
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Toni Schneider
- Institute for Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Jay Gopalakrishnan
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany.
| |
Collapse
|
16
|
Kim DS, Lee G, Cho H, Bae S. Regenerative Medicine in South Korea: Bridging the Gap Between Authorization and Reimbursement. Front Bioeng Biotechnol 2021; 9:737504. [PMID: 34527662 PMCID: PMC8435711 DOI: 10.3389/fbioe.2021.737504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/11/2021] [Indexed: 11/26/2022] Open
Abstract
Regenerative medicine (RM) has considerable potential to address the needs of aging-related and uncurable diseases. However, its incorporation into reimbursement of health insurance benefits poses many challenges, including uncertain evidence and insufficient investment. This paper examines the wide gap between manufacturers, regulatory bodies, and health technology bodies regarding reimbursements for RMs focused cell therapy products. In this mixed-methods study, we first analyzed the sales of RMs approved in South Korea. In addition to exploring beliefs related to the market value of RMs, in-depth interviews were conducted with 24 experts (17 from bio-industries, two from the regulatory body, three from a health technology assessment (HTA) body, and two from the Pharmaceutical Benefit Coverage Assessment Committee [PBCAC]). Lastly, we surveyed PBCAC members about the market value of RMs. In total, 15 of the 20 developed cell therapy products are on the market in South Korea, and amounted to 0.24% of total pharmaceutical expenditures in 2018. We identified a wide gap between stakeholders and regulators regarding the market value and pricing of RMs. The interviewees from the pharmaceutical manufacturer association raised the issue of rising manufacturing costs and proposed a specific pricing policy for RMs. To bridge the gap between approval and reimbursement, stakeholders demand an alternative framework of value-based pricing. Conditional health insurance reimbursement may be an alternative to the traditional process in order to generate evidence of the effects of RMs using “risk-based” or “outcome-based” approaches.
Collapse
Affiliation(s)
- Dong-Sook Kim
- Department of Research, Health Insurance Review and Assessment Service, Chuncheon, South Korea
| | - Geunwoo Lee
- Department of Research, Health Insurance Review and Assessment Service, Chuncheon, South Korea
| | - Hyungyung Cho
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - SeungJin Bae
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
17
|
Santos-Terra J, Deckmann I, Fontes-Dutra M, Schwingel GB, Bambini-Junior V, Gottfried C. Transcription factors in neurodevelopmental and associated psychiatric disorders: A potential convergence for genetic and environmental risk factors. Int J Dev Neurosci 2021; 81:545-578. [PMID: 34240460 DOI: 10.1002/jdn.10141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/23/2021] [Accepted: 07/02/2021] [Indexed: 12/16/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are a heterogeneous and highly prevalent group of psychiatric conditions marked by impairments in the nervous system. Their onset occurs during gestation, and the alterations are observed throughout the postnatal life. Although many genetic and environmental risk factors have been described in this context, the interactions between them challenge the understanding of the pathways associated with NDDs. Transcription factors (TFs)-a group of over 1,600 proteins that can interact with DNA, regulating gene expression through modulation of RNA synthesis-represent a point of convergence for different risk factors. In addition, TFs organize critical processes like angiogenesis, blood-brain barrier formation, myelination, neuronal migration, immune activation, and many others in a time and location-dependent way. In this review, we summarize important TF alterations in NDD and associated disorders, along with specific impairments observed in animal models, and, finally, establish hypotheses to explain how these proteins may be critical mediators in the context of genome-environment interactions.
Collapse
Affiliation(s)
- Júlio Santos-Terra
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Iohanna Deckmann
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Mellanie Fontes-Dutra
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Gustavo Brum Schwingel
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Victorio Bambini-Junior
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Preston, UK
| | - Carmem Gottfried
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| |
Collapse
|
18
|
Nalamalapu RR, Yue M, Stone AR, Murphy S, Saha MS. The tweety Gene Family: From Embryo to Disease. Front Mol Neurosci 2021; 14:672511. [PMID: 34262434 PMCID: PMC8273234 DOI: 10.3389/fnmol.2021.672511] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 12/31/2022] Open
Abstract
The tweety genes encode gated chloride channels that are found in animals, plants, and even simple eukaryotes, signifying their deep evolutionary origin. In vertebrates, the tweety gene family is highly conserved and consists of three members—ttyh1, ttyh2, and ttyh3—that are important for the regulation of cell volume. While research has elucidated potential physiological functions of ttyh1 in neural stem cell maintenance, proliferation, and filopodia formation during neural development, the roles of ttyh2 and ttyh3 are less characterized, though their expression patterns during embryonic and fetal development suggest potential roles in the development of a wide range of tissues including a role in the immune system in response to pathogen-associated molecules. Additionally, members of the tweety gene family have been implicated in various pathologies including cancers, particularly pediatric brain tumors, and neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease. Here, we review the current state of research using information from published articles and open-source databases on the tweety gene family with regard to its structure, evolution, expression during development and adulthood, biochemical and cellular functions, and role in human disease. We also identify promising areas for further research to advance our understanding of this important, yet still understudied, family of genes.
Collapse
Affiliation(s)
- Rithvik R Nalamalapu
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Michelle Yue
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Aaron R Stone
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Samantha Murphy
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Margaret S Saha
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| |
Collapse
|
19
|
Chandrasekaran V, Carta G, da Costa Pereira D, Gupta R, Murphy C, Feifel E, Kern G, Lechner J, Cavallo AL, Gupta S, Caiment F, Kleinjans JCS, Gstraunthaler G, Jennings P, Wilmes A. Generation and characterization of iPSC-derived renal proximal tubule-like cells with extended stability. Sci Rep 2021; 11:11575. [PMID: 34078926 PMCID: PMC8172841 DOI: 10.1038/s41598-021-89550-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/23/2021] [Indexed: 12/21/2022] Open
Abstract
The renal proximal tubule is responsible for re-absorption of the majority of the glomerular filtrate and its proper function is necessary for whole-body homeostasis. Aging, certain diseases and chemical-induced toxicity are factors that contribute to proximal tubule injury and chronic kidney disease progression. To better understand these processes, it would be advantageous to generate renal tissues from human induced pluripotent stem cells (iPSC). Here, we report the differentiation and characterization of iPSC lines into proximal tubular-like cells (PTL). The protocol is a step wise exposure of small molecules and growth factors, including the GSK3 inhibitor (CHIR99021), the retinoic acid receptor activator (TTNPB), FGF9 and EGF, to drive iPSC to PTL via cell stages representing characteristics of early stages of renal development. Genome-wide RNA sequencing showed that PTL clustered within a kidney phenotype. PTL expressed proximal tubular-specific markers, including megalin (LRP2), showed a polarized phenotype, and were responsive to parathyroid hormone. PTL could take up albumin and exhibited ABCB1 transport activity. The phenotype was stable for up to 7 days and was maintained after passaging. This protocol will form the basis of an optimized strategy for molecular investigations using iPSC derived PTL.
Collapse
Affiliation(s)
- Vidya Chandrasekaran
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Giada Carta
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Daniel da Costa Pereira
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Rajinder Gupta
- Department of Toxicogenomics, Maastricht University, School of Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Cormac Murphy
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Elisabeth Feifel
- Institute of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Kern
- Institute of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Judith Lechner
- Institute of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | | | | | - Florian Caiment
- Department of Toxicogenomics, Maastricht University, School of Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Jos C S Kleinjans
- Department of Toxicogenomics, Maastricht University, School of Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Gerhard Gstraunthaler
- Institute of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands.
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands.
| |
Collapse
|
20
|
From Proteomic Mapping to Invasion-Metastasis-Cascade Systemic Biomarkering and Targeted Drugging of Mutant BRAF-Dependent Human Cutaneous Melanomagenesis. Cancers (Basel) 2021; 13:cancers13092024. [PMID: 33922182 PMCID: PMC8122743 DOI: 10.3390/cancers13092024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/09/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Despite the recent advances in human malignancy therapy, metastasis and chemoresistance remain the principal causes of cancer-derived deaths. Given the fatal forms of cutaneous metastatic melanoma, we herein employed primary (WM115) and metastatic (WM266-4) melanoma cells, both obtained from the same patient, to identify novel biomarkers and therapeutic agents. Through state-of-the-art technologies including deep proteome landscaping, immunofluorescence phenotyping, and drug toxicity screening, we were able to describe new molecular programs, oncogenic drivers, and drug regimens, controlling the invasion-metastasis cascade during BRAFV600D-dependent melanomagenesis. It proved that proteomic navigation could foster the development of systemic biomarkering and targeted drugging for successful treatment of advanced disease. Abstract Melanoma is classified among the most notoriously aggressive human cancers. Despite the recent progress, due to its propensity for metastasis and resistance to therapy, novel biomarkers and oncogenic molecular drivers need to be promptly identified for metastatic melanoma. Hence, by employing nano liquid chromatography-tandem mass spectrometry deep proteomics technology, advanced bioinformatics algorithms, immunofluorescence, western blotting, wound healing protocols, molecular modeling programs, and MTT assays, we comparatively examined the respective proteomic contents of WM115 primary (n = 3955 proteins) and WM266-4 metastatic (n = 6681 proteins) melanoma cells. It proved that WM115 and WM266-4 cells have engaged hybrid epithelial-to-mesenchymal transition/mesenchymal-to-epithelial transition states, with TGF-β controlling their motility in vitro. They are characterized by different signatures of SOX-dependent neural crest-like stemness and distinct architectures of the cytoskeleton network. Multiple signaling pathways have already been activated from the primary melanoma stage, whereas HIF1α, the major hypoxia-inducible factor, can be exclusively observed in metastatic melanoma cells. Invasion-metastasis cascade-specific sub-routines of activated Caspase-3-triggered apoptosis and LC3B-II-dependent constitutive autophagy were also unveiled. Importantly, WM115 and WM266-4 cells exhibited diverse drug response profiles, with epirubicin holding considerable promise as a beneficial drug for metastatic melanoma clinical management. It is the proteome navigation that enables systemic biomarkering and targeted drugging to open new therapeutic windows for advanced disease.
Collapse
|
21
|
Foss-Skiftesvik J, Hagen CM, Mathiasen R, Adamsen D, Bækvad-Hansen M, Børglum AD, Nordentoft M, Werge T, Christiansen M, Schmiegelow K, Juhler M, Mortensen PB, Hougaard DM, Bybjerg-Grauholm J. Genome-wide association study across pediatric central nervous system tumors implicates shared predisposition and points to 1q25.2 (PAPPA2) and 11p12 (LRRC4C) as novel candidate susceptibility loci. Childs Nerv Syst 2021; 37:819-830. [PMID: 33226468 DOI: 10.1007/s00381-020-04946-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/26/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Central nervous system (CNS) tumors constitute the most common form of solid neoplasms in children, but knowledge on genetic predisposition is sparse. In particular, whether susceptibility attributable to common variants is shared across CNS tumor types in children has not been investigated. The purpose of this study was to explore potential common genetic risk variants exhibiting pleiotropic effects across pediatric CNS tumors. We also investigated whether such susceptibility differs between early and late onset of disease. METHOD A Danish nationwide genome-wide association study (GWAS) of 1,097 consecutive patients (< 15 years of age) with CNS tumors and a cohort of 4,745 population-based controls. RESULTS For both the overall cohort and patients diagnosed after the age of four, the strongest association was rs12064625 which maps to PAPPA2 at 1q25.2 (p = 3.400 × 10-7 and 9.668 × 10-8, respectively). PAPPA2 regulates local bioavailability of insulin-like growth factor I (IGF-I). IGF-I is fundamental to CNS development and is involved in tumorigenesis across a wide range of different cancers. For the younger children, the strongest association was provided by rs11036373 mapping to LRRC4C at 11p12 (p = 7.620 × 10-7), which encoded protein acts as an axon guidance molecule during CNS development and has not formerly been associated with brain tumors. DISCUSSION This GWAS indicates shared susceptibility attributable to common variants across pediatric CNS tumor types. Variations in genetic loci with roles in CNS development appear to be involved, possibly via altered IGF-I related pathways.
Collapse
Affiliation(s)
- Jon Foss-Skiftesvik
- Department of Neurosurgery, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark.
- Danish Center for Neonatal Screening, Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark.
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet University Hospital, Copenhagen, Denmark.
| | - Christian Munch Hagen
- Danish Center for Neonatal Screening, Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
| | - René Mathiasen
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet University Hospital, Copenhagen, Denmark
| | - Dea Adamsen
- Danish Center for Neonatal Screening, Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
| | - Marie Bækvad-Hansen
- Danish Center for Neonatal Screening, Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
- Department of Biomedicine, Aarhus University and Centre for Integrative Sequencing, iSEQ, Aarhus, Denmark
- Aarhus Genome Center, Aarhus, Denmark
| | - Merete Nordentoft
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
- Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas Werge
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
| | - Michael Christiansen
- Danish Center for Neonatal Screening, Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet University Hospital, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marianne Juhler
- Department of Neurosurgery, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - Preben Bo Mortensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
- National Centre for Register-based Research, Department of Economics and Business Economics, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-Based Research, Aarhus University, Aarhus, Denmark
| | - David Michael Hougaard
- Danish Center for Neonatal Screening, Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
| | - Jonas Bybjerg-Grauholm
- Danish Center for Neonatal Screening, Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen, Denmark
| |
Collapse
|
22
|
Cabau-Laporta J, Ascensión AM, Arrospide-Elgarresta M, Gerovska D, Araúzo-Bravo MJ. FOntCell: Fusion of Ontologies of Cells. Front Cell Dev Biol 2021; 9:562908. [PMID: 33644039 PMCID: PMC7905052 DOI: 10.3389/fcell.2021.562908] [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: 05/18/2020] [Accepted: 01/05/2021] [Indexed: 11/25/2022] Open
Abstract
High-throughput cell-data technologies such as single-cell RNA-seq create a demand for algorithms for automatic cell classification and characterization. There exist several cell classification ontologies with complementary information. However, one needs to merge them to synergistically combine their information. The main difficulty in merging is to match the ontologies since they use different naming conventions. Therefore, we developed an algorithm that merges ontologies by integrating the name matching between class label names with the structure mapping between the ontology elements based on graph convolution. Since the structure mapping is a time consuming process, we designed two methods to perform the graph convolution: vectorial structure matching and constraint-based structure matching. To perform the vectorial structure matching, we designed a general method to calculate the similarities between vectors of different lengths for different metrics. Additionally, we adapted the slower Blondel method to work for structure matching. We implemented our algorithms into FOntCell, a software module in Python for efficient automatic parallel-computed merging/fusion of ontologies in the same or similar knowledge domains. FOntCell can unify dispersed knowledge from one domain into a unique ontology in OWL format and iteratively reuse it to continuously adapt ontologies with new data endlessly produced by data-driven classification methods, such as of the Human Cell Atlas. To navigate easily across the merged ontologies, it generates HTML files with tabulated and graphic summaries, and interactive circular Directed Acyclic Graphs. We used FOntCell to merge the CELDA, LifeMap and LungMAP Human Anatomy cell ontologies into a comprehensive cell ontology. We compared FOntCell with tools used for the alignment of mouse and human anatomy ontologies task proposed by the Ontology Alignment Evaluation Initiative (OAEI) and found that the Fβ alignment accuracies of FOntCell are above the geometric mean of the other tools; more importantly, it outperforms significantly the best OAEI tools in cell ontology alignment in terms of Fβ alignment accuracies.
Collapse
Affiliation(s)
- Javier Cabau-Laporta
- Computational Biology and Systems Biomedicine Group, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Alex M Ascensión
- Computational Biology and Systems Biomedicine Group, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Mikel Arrospide-Elgarresta
- Computational Biology and Systems Biomedicine Group, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Daniela Gerovska
- Computational Biology and Systems Biomedicine Group, Biodonostia Health Research Institute, San Sebastián, Spain.,Computational Biomedicine Data Analysis Platform, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine Group, Biodonostia Health Research Institute, San Sebastián, Spain.,Computational Biomedicine Data Analysis Platform, Biodonostia Health Research Institute, San Sebastián, Spain.,Basque Foundation for Science (IKERBASQUE), Bilbao, Spain.,Centro de Investigación Biomédica en Red (CIBER) of Frailty and Healthy Aging (CIBERfes), Madrid, Spain.,TransBioNet Thematic Network of Excellence for Transitional Bioinformatics, Barcelona Supercomputing Center, Barcelona, Spain.,Computational Biology and Bioinformatics, Department Cell and Developmental Biology Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
23
|
Yu Z, Zou H, Wang H, Li Q, Yu D. Identification of Key Gene Signatures Associated With Bone Metastasis in Castration-Resistant Prostate Cancer Using Co-Expression Analysis. Front Oncol 2021; 10:571524. [PMID: 33604283 PMCID: PMC7884857 DOI: 10.3389/fonc.2020.571524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 12/14/2020] [Indexed: 11/13/2022] Open
Abstract
About 80–90% of castration-resistant prostate cancer (CRPC) patients would develop bone metastasis. However, the molecular mechanisms of bone metastasis are still not clear. This study aimed to detect the differences between the tumor and normal samples in bone after metastatic colonization. Four transcriptional datasets (GSE32269, GSE101607, GSE29650, and GSE74685) were obtained from the GEO database. 1983 differentially expressed genes (DEGs) were first identified between tumor and normal marrow samples in GSE32269. Most of the top 10 up-regulated DEGs are related with prostate cancer, and the top 10 down-regulated DEGs are mainly related with bone development. Seven co-expression modules were then detected based on the 1469 DEGs shared by the four datasets. Three of them were found highly preserved among the four datasets. Enrichment analysis showed that the three modules were respectively enriched in Cell adhesion molecules (CAMs), Leukocyte transendothelial migration and cell cycle, which might play significantly important roles in the tumor development in bone marrow. Ten, 17, and 99 hub genes for each module were then identified. And four genes (C3AR1, IL10RA, LY86, and MS4A6A) were detect to be tightly related to progression of bone metastatic CRPC. ROC curve was plotted and AUC was calculated to distinguish tumor and normal bone marrow samples as well as bone and non-bone metastatic CRPCs. The present study identified key genes and modules involved in bone metastatic CRPCs, which may provide new insights and biomarkers for understanding of the molecular mechanisms of bone metastatic CRPC.
Collapse
Affiliation(s)
- Zhongxiang Yu
- Department of Orthopaedics, Shuguang Hospital Affiliated to Shanghai Traditional Chinese Medical University, Shanghai, China
| | - Hanlin Zou
- Department of Orthopedics, Putuo Hospital Affiliated to Shanghai Traditional Chinese Medical University, Shanghai, China
| | - Huihao Wang
- Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai Traditional Chinese Medical University, Shanghai, China
| | - Qi Li
- Department of Oncology, Shuguang Hospital Affiliated to Shanghai Traditional Chinese Medical University, Shanghai, China
| | - Dong Yu
- Center for Translational Medicine, Second Military Medical University, Shanghai, China
| |
Collapse
|
24
|
Burton J, Umu SU, Langseth H, Grotmol T, Grimsrud TK, Haugen TB, Rounge TB. Serum RNA Profiling in the 10-Years Period Prior to Diagnosis of Testicular Germ Cell Tumor. Front Oncol 2020; 10:574977. [PMID: 33251139 PMCID: PMC7673397 DOI: 10.3389/fonc.2020.574977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Although testicular germ cell tumor (TGCT) overall is highly curable, patients may experience late effects after treatment. An increased understanding of the mechanisms behind the development of TGCT may pave the way for better outcome for patients. To elucidate molecular changes prior to TGCT diagnosis we sequenced small RNAs in serum from 69 patients who were later diagnosed with TGCT and 111 matched controls. The deep RNA profiles, with on average 18 million sequences per sample, comprised of nine classes of RNA, including microRNA. We found that circulating RNA signals differed significantly between cases and controls regardless of time to diagnosis. Different levels of TSIX related to X-chromosome inactivation and TEX101 involved in spermatozoa production are among the interesting findings. The RNA signals differed between seminoma and non-seminoma TGCT subtypes, with seminoma cases showing lower levels of RNAs and non-seminoma cases showing higher levels of RNAs, compared with controls. The differentially expressed RNAs were typically associated with cancer related pathways. Our results indicate that circulating RNA profiles change during TGCT development according to histology and may be useful for early detection of this tumor type.
Collapse
Affiliation(s)
- Joshua Burton
- Department of Lifesciences and Health, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Sinan U. Umu
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Hilde Langseth
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Tom Grotmol
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Tom K. Grimsrud
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Trine B. Haugen
- Department of Lifesciences and Health, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Trine B. Rounge
- Department of Research, Cancer Registry of Norway, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| |
Collapse
|
25
|
Mota C, Camarero-Espinosa S, Baker MB, Wieringa P, Moroni L. Bioprinting: From Tissue and Organ Development to in Vitro Models. Chem Rev 2020; 120:10547-10607. [PMID: 32407108 PMCID: PMC7564098 DOI: 10.1021/acs.chemrev.9b00789] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Indexed: 02/08/2023]
Abstract
Bioprinting techniques have been flourishing in the field of biofabrication with pronounced and exponential developments in the past years. Novel biomaterial inks used for the formation of bioinks have been developed, allowing the manufacturing of in vitro models and implants tested preclinically with a certain degree of success. Furthermore, incredible advances in cell biology, namely, in pluripotent stem cells, have also contributed to the latest milestones where more relevant tissues or organ-like constructs with a certain degree of functionality can already be obtained. These incredible strides have been possible with a multitude of multidisciplinary teams around the world, working to make bioprinted tissues and organs more relevant and functional. Yet, there is still a long way to go until these biofabricated constructs will be able to reach the clinics. In this review, we summarize the main bioprinting activities linking them to tissue and organ development and physiology. Most bioprinting approaches focus on mimicking fully matured tissues. Future bioprinting strategies might pursue earlier developmental stages of tissues and organs. The continuous convergence of the experts in the fields of material sciences, cell biology, engineering, and many other disciplines will gradually allow us to overcome the barriers identified on the demanding path toward manufacturing and adoption of tissue and organ replacements.
Collapse
Affiliation(s)
- Carlos Mota
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Sandra Camarero-Espinosa
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Matthew B. Baker
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Paul Wieringa
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| |
Collapse
|
26
|
Sturm G, Finotello F, Petitprez F, Zhang JD, Baumbach J, Fridman WH, List M, Aneichyk T. Comprehensive evaluation of transcriptome-based cell-type quantification methods for immuno-oncology. Bioinformatics 2020; 35:i436-i445. [PMID: 31510660 PMCID: PMC6612828 DOI: 10.1093/bioinformatics/btz363] [Citation(s) in RCA: 517] [Impact Index Per Article: 129.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MOTIVATION The composition and density of immune cells in the tumor microenvironment (TME) profoundly influence tumor progression and success of anti-cancer therapies. Flow cytometry, immunohistochemistry staining or single-cell sequencing are often unavailable such that we rely on computational methods to estimate the immune-cell composition from bulk RNA-sequencing (RNA-seq) data. Various methods have been proposed recently, yet their capabilities and limitations have not been evaluated systematically. A general guideline leading the research community through cell type deconvolution is missing. RESULTS We developed a systematic approach for benchmarking such computational methods and assessed the accuracy of tools at estimating nine different immune- and stromal cells from bulk RNA-seq samples. We used a single-cell RNA-seq dataset of ∼11 000 cells from the TME to simulate bulk samples of known cell type proportions, and validated the results using independent, publicly available gold-standard estimates. This allowed us to analyze and condense the results of more than a hundred thousand predictions to provide an exhaustive evaluation across seven computational methods over nine cell types and ∼1800 samples from five simulated and real-world datasets. We demonstrate that computational deconvolution performs at high accuracy for well-defined cell-type signatures and propose how fuzzy cell-type signatures can be improved. We suggest that future efforts should be dedicated to refining cell population definitions and finding reliable signatures. AVAILABILITY AND IMPLEMENTATION A snakemake pipeline to reproduce the benchmark is available at https://github.com/grst/immune_deconvolution_benchmark. An R package allows the community to perform integrated deconvolution using different methods (https://grst.github.io/immunedeconv). SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Gregor Sturm
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.,Pieris Pharmaceuticals GmbH, Freising, Germany
| | - Francesca Finotello
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Florent Petitprez
- Cordeliers Research Centre, UMRS_1138, INSERM, University Paris-Descartes, Sorbonne University, Paris, France.,Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Jitao David Zhang
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jan Baumbach
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Wolf H Fridman
- Cordeliers Research Centre, UMRS_1138, INSERM, University Paris-Descartes, Sorbonne University, Paris, France
| | - Markus List
- Big Data in BioMedicine Group, Chair of Experimental Bioinformatis, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Tatsiana Aneichyk
- Pieris Pharmaceuticals GmbH, Freising, Germany.,Independent Data Lab UG, Munich, Germany
| |
Collapse
|
27
|
Fracaro L, Senegaglia AC, Herai RH, Leitolis A, Boldrini-Leite LM, Rebelatto CLK, Travers PJ, Brofman PRS, Correa A. The Expression Profile of Dental Pulp-Derived Stromal Cells Supports Their Limited Capacity to Differentiate into Adipogenic Cells. Int J Mol Sci 2020; 21:E2753. [PMID: 32326648 PMCID: PMC7215853 DOI: 10.3390/ijms21082753] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/26/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) can self-renew, differentiate into specialised cells and have different embryonic origins-ectodermal for dental pulp-derived MSCs (DPSCs) and mesodermal for adipose tissue-derived MSCs (ADSCs). Data on DPSCs adipogenic differentiation potential and timing vary, and the lack of molecular and genetic information prompted us to gain a better understanding of DPSCs adipogenic differentiation potential and gene expression profile. While DPSCs differentiated readily along osteogenic and chondrogenic pathways, after 21 days in two different types of adipogenic induction media, DPSCs cultures did not contain lipid vacuoles and had low expression levels of the adipogenic genes proliferator-activated receptor gamma (PPARG), lipoprotein lipase (LPL) and CCAAT/enhancer-binding protein alpha (CEBPA). To better understand this limitation in adipogenesis, transcriptome analysis in undifferentiated DPSCs was carried out, with the ADSC transcriptome used as a positive control. In total, 14,871 transcripts were common to DPSCs and ADSCs, some were unique (DPSCs: 471, ADSCs: 1032), and 510 were differentially expressed genes. Detailed analyses of overrepresented transcripts showed that DPSCs express genes that inhibit adipogenic differentiation, revealing the possible mechanism for their limited adipogenesis.
Collapse
Affiliation(s)
- Letícia Fracaro
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná—PUCPR, Curitiba, Parana 80215-901, Brazil; (L.F.); (A.C.S.); (L.M.B.-L.); (C.L.K.R.)
| | - Alexandra C. Senegaglia
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná—PUCPR, Curitiba, Parana 80215-901, Brazil; (L.F.); (A.C.S.); (L.M.B.-L.); (C.L.K.R.)
| | - Roberto H. Herai
- Graduate Program in Health Sciences (PPGCS), School of Medicine, Pontifícia Universidade Católica do Paraná—PUCPR, Curitiba, Parana 80215-901, Brazil;
| | - Amanda Leitolis
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Fiocruz-Parana, Curitiba, Parana 81350-010, Brazil;
| | - Lidiane M. Boldrini-Leite
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná—PUCPR, Curitiba, Parana 80215-901, Brazil; (L.F.); (A.C.S.); (L.M.B.-L.); (C.L.K.R.)
| | - Carmen L. K. Rebelatto
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná—PUCPR, Curitiba, Parana 80215-901, Brazil; (L.F.); (A.C.S.); (L.M.B.-L.); (C.L.K.R.)
| | - Paul J. Travers
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, Scotland, UK;
| | - Paulo R. S. Brofman
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná—PUCPR, Curitiba, Parana 80215-901, Brazil; (L.F.); (A.C.S.); (L.M.B.-L.); (C.L.K.R.)
| | - Alejandro Correa
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Fiocruz-Parana, Curitiba, Parana 81350-010, Brazil;
| |
Collapse
|
28
|
Bi Y, Tu Z, Zhang Y, Yang P, Guo M, Zhu X, Zhao C, Zhou J, Wang H, Wang Y, Gao S. Identification of ALPPL2 as a Naive Pluripotent State-Specific Surface Protein Essential for Human Naive Pluripotency Regulation. Cell Rep 2020; 30:3917-3931.e5. [DOI: 10.1016/j.celrep.2020.02.090] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/28/2019] [Accepted: 02/25/2020] [Indexed: 10/24/2022] Open
|
29
|
Finkelstein J, Parvanova I, Zhang F. Informatics Approaches for Harmonized Intelligent Integration of Stem Cell Research. Stem Cells Cloning 2020; 13:1-20. [PMID: 32099411 PMCID: PMC6996484 DOI: 10.2147/sccaa.s237361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/11/2020] [Indexed: 12/15/2022] Open
Abstract
As biomedical data integration and analytics play an increasing role in the field of stem cell research, it becomes important to develop ways to standardize, aggregate, and share data among researchers. For this reason, many databases have been developed in recent years in an attempt to systematically warehouse data from different stem cell projects and experiments at the same time. However, these databases vary widely in their implementation and structure. The aim of this scoping review is to characterize the main features of available stem cell databases in order to identify specifications useful for implementation in future stem cell databases. We conducted a scoping review of peer-reviewed literature and online resources to identify and review available stem cell databases. To identify the relevant databases, we performed a PubMed search using relevant MeSH terms followed by a web search for databases which may not have an associated journal article. In total, we identified 16 databases to include in this review. The data elements reported in these databases represented a broad spectrum of parameters from basic socio-demographic variables to various cells characteristics, cell surface markers expression, and clinical trial results. Three broad sets of functional features that provide utility for future stem cell research and facilitate bioinformatics workflows were identified. These features consisted of the following: common data elements, data visualization and analysis tools, and biomedical ontologies for data integration. Stem cell bioinformatics is a quickly evolving field that generates a growing number of heterogeneous data sets. Further progress in the stem cell research may be greatly facilitated by development of applications for intelligent stem cell data aggregation, sharing and collaboration process.
Collapse
Affiliation(s)
- Joseph Finkelstein
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Irena Parvanova
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Frederick Zhang
- Center for Bioinformatics and Data Analytics, Columbia University, New York, NY, USA
| |
Collapse
|
30
|
O'Donnell L, Blakely EL, Baty K, Alexander M, Bogdanova-Mihaylova P, Craig J, Walsh R, Brett F, Taylor RW, Murphy SM. Chronic Progressive External Ophthalmoplegia due to a Rare de novo m.12334G>A MT-TL2 Mitochondrial DNA Variant1. J Neuromuscul Dis 2020; 7:355-360. [PMID: 32310184 DOI: 10.3233/jnd-200486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We describe a patient with chronic progressive external ophthalmoplegia (CPEO) due to a rare mitochondrial genetic variant. Muscle biopsy revealed numerous cytochrome c oxidase (COX)-deficient fibres, prompting sequencing of the entire mitochondrial genome in muscle which revealed a rare m.12334G>A variant in the mitochondrial (mt-) tRNALeu(CUN)(MT-TL2) gene. Analysis of several tissues showed this to be a de novo mutational event. Single fibre studies confirmed the segregation of high m.12334G>A heteroplasmy levels with the COX histochemical defect, confirming pathogenicity of the m.12334G>A MT-TL2 variant. This case illustrates the importance of pursuing molecular genetic analysis in clinically-affected tissues when mitochondrial disease is suspected.
Collapse
Affiliation(s)
- Luke O'Donnell
- Department of Neurology, Tallaght University Hospital, Tallaght, Dublin, Ireland
| | - Emma L Blakely
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- NHS Highly Specialised Mitochondrial Diagnostic Laboratory, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Karen Baty
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- NHS Highly Specialised Mitochondrial Diagnostic Laboratory, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Michael Alexander
- Department of Neurophysiology, Tallaght University Hospital, Tallaght, Dublin, Ireland
- Academic Unit of Neurology, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | | | - John Craig
- Department of Neurology, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Ronan Walsh
- Department of Neurology, Hermitage Medical Clinic, Dublin, Ireland
| | - Francesca Brett
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- NHS Highly Specialised Mitochondrial Diagnostic Laboratory, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Sinead M Murphy
- Department of Neurology, Tallaght University Hospital, Tallaght, Dublin, Ireland
- Academic Unit of Neurology, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| |
Collapse
|
31
|
Rojo C, Zhang Q, Keleş S. iFunMed: Integrative functional mediation analysis of GWAS and eQTL studies. Genet Epidemiol 2019; 43:742-760. [PMID: 31328826 DOI: 10.1002/gepi.22217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/17/2019] [Accepted: 05/07/2019] [Indexed: 11/08/2022]
Abstract
Genome-wide association studies (GWAS) have successfully identified thousands of genetic variants contributing to disease and other phenotypes. However, significant obstacles hamper our ability to elucidate causal variants, identify genes affected by causal variants, and characterize the mechanisms by which genotypes influence phenotypes. The increasing availability of genome-wide functional annotation data is providing unique opportunities to incorporate prior information into the analysis of GWAS to better understand the impact of variants on disease etiology. Although there have been many advances in incorporating prior information into prioritization of trait-associated variants in GWAS, functional annotation data have played a secondary role in the joint analysis of GWAS and molecular (i.e., expression) quantitative trait loci (eQTL) data in assessing evidence for association. To address this, we develop a novel mediation framework, iFunMed, to integrate GWAS and eQTL data with the utilization of publicly available functional annotation data. iFunMed extends the scope of standard mediation analysis by incorporating information from multiple genetic variants at a time and leveraging variant-level summary statistics. Data-driven computational experiments convey how informative annotations improve single-nucleotide polymorphism (SNP) selection performance while emphasizing robustness of iFunMed to noninformative annotations. Application to Framingham Heart Study data indicates that iFunMed is able to boost detection of SNPs with mediation effects that can be attributed to regulatory mechanisms.
Collapse
Affiliation(s)
- Constanza Rojo
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Qi Zhang
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Sündüz Keleş
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
32
|
Pawar G, Madden JC, Ebbrell D, Firman JW, Cronin MTD. In Silico Toxicology Data Resources to Support Read-Across and (Q)SAR. Front Pharmacol 2019; 10:561. [PMID: 31244651 PMCID: PMC6580867 DOI: 10.3389/fphar.2019.00561] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022] Open
Abstract
A plethora of databases exist online that can assist in in silico chemical or drug safety assessment. However, a systematic review and grouping of databases, based on purpose and information content, consolidated in a single source, has been lacking. To resolve this issue, this review provides a comprehensive listing of the key in silico data resources relevant to: chemical identity and properties, drug action, toxicology (including nano-material toxicity), exposure, omics, pathways, Absorption, Distribution, Metabolism and Elimination (ADME) properties, clinical trials, pharmacovigilance, patents-related databases, biological (genes, enzymes, proteins, other macromolecules etc.) databases, protein-protein interactions (PPIs), environmental exposure related, and finally databases relating to animal alternatives in support of 3Rs policies. More than nine hundred databases were identified and reviewed against criteria relating to accessibility, data coverage, interoperability or application programming interface (API), appropriate identifiers, types of in vitro, in vivo,-clinical or other data recorded and suitability for modelling, read-across, or similarity searching. This review also specifically addresses the need for solutions for mapping and integration of databases into a common platform for better translatability of preclinical data to clinical data.
Collapse
Affiliation(s)
| | | | | | | | - Mark T. D. Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
33
|
Shen Q, Yu S, Zhang Y, Zhou Z, Zhu Z, Pan Q, Lv S, Niu H, Li N, Peng S, Liao M, Wang H, Lei A, Miao Y, Liu Z, Hua J. Characterization of porcine extraembryonic endoderm cells. Cell Prolif 2019; 52:e12591. [PMID: 30896067 PMCID: PMC6536407 DOI: 10.1111/cpr.12591] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES To date, many efforts have been made to establish porcine embryonic stem (pES) cells without success. Extraembryonic endoderm (XEN) cells can self-renew and differentiate into the visceral endoderm and parietal endoderm. XEN cells are derived from the primitive endoderm of the inner cell mass of blastocysts and may be an intermediate state in cell reprogramming. MATERIALS AND METHODS Porcine XEN cells (pXENCs) were generated from porcine pluripotent stem cells (pPSCs) and were characterized by RNA sequencing and immunofluorescence analyses. The developmental potential of pXENCs was investigated in chimeric mouse embryos. RESULTS Porcine XEN cells derived from porcine pPSCs were successfully expanded in N2B27 medium supplemented with bFGF for least 30 passages. RNA sequencing and immunofluorescence analyses showed that pXENCs expressed the murine and canine XEN markers Gata6, Gata4, Sox17 and Pdgfra but not the pluripotent markers Oct4, Sox2 and TE marker Cdx2. Moreover, these cells contributed to the XEN when injected into four-cell stage mouse embryos. Supplementation with Chir99021 and SB431542 promoted the pluripotency of the pXENCs. CONCLUSIONS We successfully derived pXENCs and showed that supplementation with Chir99021 and SB431542 confer them with pluripotency. Our results provide a new resource for investigating the reprogramming mechanism of porcine-induced pluripotent stem cells.
Collapse
Affiliation(s)
- Qiao‐Yan Shen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Shuai Yu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Ying Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Zhe Zhou
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Zhen‐Shuo Zhu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Qin Pan
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Shan Lv
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Hui‐Min Niu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Ming‐zhi Liao
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Hua‐Yan Wang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - An‐Min Lei
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| | - Yi‐Liang Miao
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Zhong‐Hua Liu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life ScienceNorth‐East Agricultural UniversityHarbinChina
| | - Jin‐Lian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and TechnologyNorthwest A&F UniversityYanglingChina
| |
Collapse
|
34
|
Gianni C, Sanchez JE, Mohanty S, Trivedi C, Della Rocca DG, Al-Ahmad A, Burkhardt JD, Gallinghouse GJ, Hranitzky PM, Horton RP, Di Biase L, Natale A. Isolation of the superior vena cava from the right atrial posterior wall: a novel ablation approach. Europace 2019; 20:e124-e132. [PMID: 29016788 DOI: 10.1093/europace/eux262] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/10/2017] [Indexed: 11/14/2022] Open
Abstract
Aims Superior vena cava (SVC) isolation might be difficult to achieve because of the vicinity of the phrenic nerve (PN) and sinus node. Based on its embryogenesis, we hypothesized the presence of preferential conduction from the right atrial (RA) posterior wall, making it possible to isolate the SVC antrally, sparing its anterior and lateral aspect. Methods and results This is a descriptive cohort study of 105 consecutive patients in which SVC isolation was obtained with radiofrequency ablation, starting in the septal aspect of the SVC-RA junction and continued posteriorly and inferiorly targeting sites of early activation until electrical isolation was obtained. Acute SVC isolation was achieved in 103 (98%) patients; the mean distance between the site of SVC isolation and the SVC-RA junction was 19.9 ± 5.3 (range 9.7-33.7) mm. During follow-up, 2 (2%) patients developed symptomatic diaphragmatic paralysis due to transient right PN injury; 13 patients underwent a repeat ablation: SVC reconnection was observed in 5 patients, and re-isolation was easily achieved by targeting the corresponding sites of early activation. Conclusion Superior vena cava isolation can be completed by targeting its septal segment and sites of early activation in the posterior SVC-RA junction and RA posterior wall; this is a feasible alternative ablation strategy in patients in which SVC isolation cannot be completed with the standard approach. The risk of sinus node injury or SVC stenosis are eliminated; PN injury is still possible but can easily be prevented with high-output pacing to exclude a true posterior course of the PN.
Collapse
Affiliation(s)
- Carola Gianni
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA
| | - Javier E Sanchez
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA
| | - Sanghamitra Mohanty
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA.,Dell Medical School, University of Texas, Austin, TX, USA
| | - Chintan Trivedi
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA
| | - Domenico G Della Rocca
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA.,Department of Cardiology, University of Tor Vergata, Rome, Italy
| | - Amin Al-Ahmad
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA
| | - J David Burkhardt
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA
| | - G Joseph Gallinghouse
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA
| | - Patrick M Hranitzky
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA
| | - Rodney P Horton
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA.,Department of Biomedical Engineering, University of Texas, Austin, TX, USA
| | - Luigi Di Biase
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA.,Department of Biomedical Engineering, University of Texas, Austin, TX, USA.,Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Andrea Natale
- Texas Cardiac Arrhythmia Institute, St. David's Medical Center, 3000 N IH-35, Suite 720, Austin, TX, USA.,Dell Medical School, University of Texas, Austin, TX, USA.,Department of Biomedical Engineering, University of Texas, Austin, TX, USA.,Interventional Electrophysiology, Scripps Clinic, La Jolla, CA, USA.,Metro Health Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Division of Cardiology, Stanford University, Stanford, CA, USA.,Electrophysiology and Arrhythmia Services, California Pacific Medical Center, San Francisco, CA, USA
| |
Collapse
|
35
|
Shin J, Rhim J, Kwon Y, Choi SY, Shin S, Ha CW, Lee C. Comparative analysis of differentially secreted proteins in serum-free and serum-containing media by using BONCAT and pulsed SILAC. Sci Rep 2019; 9:3096. [PMID: 30816242 PMCID: PMC6395664 DOI: 10.1038/s41598-019-39650-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 01/29/2019] [Indexed: 01/15/2023] Open
Abstract
Despite the increased interest in secretomes associated with paracrine/autocrine mechanisms, the majority of mass spectrometric cell secretome studies have been performed using serum-free medium (SFM). On the other hand, serum-containing medium (SCM) is not recommended very much because the secretome obtained with SCM is easily contaminated with fetal bovine serum (FBS) proteins. In this study, through the combination of bioorthogonal non-canonical amino acid tagging (BONCAT) and pulsed-SILAC (pSILAC), we analyzed differentially secreted proteins between SFM and SCM in a cancer-derived human cell, U87MG, and a mesenchymal stem cell derived from human Wharton’s jelly (hWJ-MSCs). In most cases, the bioinformatic tools predicted a protein to be truly secretory when the secretion level of the protein was more in SCM than in SFM. In the case of hWJ-MSCs, the amount of proteins secreted in SCM for 24 hours was larger than that of SFM (log2 fold change = 0.96), even considering different cell proliferation rates. hWJ-MSCs proteins secreted more in SCM included several positive markers of MSC paracrine factors implicated in angiogenesis, neurogenesis and osteogenesis, and upstream regulators of cell proliferation. Our study suggests the analysis of the secretome should be processed in SCM that promotes cell proliferation and secretion.
Collapse
Affiliation(s)
- Jihye Shin
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul, 02792, Korea.,Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, 8 Kanazawa, Yokohama, 236-0004, Japan
| | - Jiheon Rhim
- Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, Korea
| | - Yumi Kwon
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul, 02792, Korea.,Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Sun Young Choi
- Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
| | - Sungho Shin
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul, 02792, Korea.,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Korea
| | - Chul-Won Ha
- Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
| | - Cheolju Lee
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul, 02792, Korea. .,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Korea. .,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Korea.
| |
Collapse
|
36
|
West MD, Chang CF, Larocca D, Li J, Jiang J, Sim P, Labat I, Chapman KB, Wong KE, Nicoll J, Van Kanegan MJ, de Grey ADNJ, Nasonkin IO, Stahl A, Sternberg H. Clonal derivation of white and brown adipocyte progenitor cell lines from human pluripotent stem cells. Stem Cell Res Ther 2019; 10:7. [PMID: 30616682 PMCID: PMC6323697 DOI: 10.1186/s13287-018-1087-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/10/2018] [Accepted: 11/21/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The role of brown fat in non-shivering thermogenesis and the discovery of brown fat depots in adult humans has made it the subject of intense research interest. A renewable source of brown adipocyte (BA) progenitors would be highly valuable for research and therapy. Directed differentiation of human pluripotent stem (hPS) cells to white or brown adipocytes is limited by lack of cell purity and scalability. Here we describe an alternative approach involving the identification of clonal self-renewing human embryonic progenitor (hEP) cell lines following partial hPS cell differentiation and selection of scalable clones. METHODS We screened a diverse panel of hPS cell-derived clonal hEP cell lines for adipocyte markers following growth in adipocyte differentiation medium. The transcriptome of the human hES-derived clonal embryonic progenitor cell lines E3, C4ELS5.1, NP88, and NP110 representing three class of definitive adipocyte progenitors were compared to the relatively non-adipogenic line E85 and adult-derived BAT and SAT-derived cells using gene expression microarrays, RT-qPCR, metabolic analysis and immunocytochemistry. Differentiation conditions were optimized for maximal UCP1 expression. RESULTS Many of the differentiated hEP cell lines expressed the adipocyte marker, FAPB4, but only a small subset expressed definitive adipocyte markers including brown adipocyte marker, UCP1. Class I cells (i.e., E3) expressed CITED1, ADIPOQ, and C19orf80 but little to no UCP1. Class II (i.e., C4ELS5.1) expressed CITED1 and UCP1 but little ADIPOQ and LIPASIN. Class III (i.e., NP88, NP110) expressed CITED1, ADIPOQ, C19orf80, and UCP1 in a similar manner as fetal BAT-derived (fBAT) cells. Differentiated NP88 and NP110 lines were closest to fBAT cells morphologically in adiponectin and uncoupling protein expression. But they were more metabolically active than fBAT cells, had higher levels of 3-hydroxybutyrate, and lacked expression of fetal/adult marker, COX7A1. The hEP BA progenitor lines were scalable to 17 passages without loss of differentiation capacity and could be readily rederived. CONCLUSIONS Taken together, these data demonstrate that self-renewing adipocyte progenitor cells can be derived from hES cells and that they are functionally like BAT cells but with unique properties that might be advantageous for basic research and for development of cell-based treatments for metabolic diseases.
Collapse
Affiliation(s)
- Michael D. West
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Ching-Fang Chang
- 0000 0001 2181 7878grid.47840.3fUniversity of California, Berkeley, CA 94720 USA
| | - Dana Larocca
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Jie Li
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Jianjie Jiang
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Pamela Sim
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Ivan Labat
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Karen B. Chapman
- 0000 0001 2171 9311grid.21107.35Johns Hopkins University, Baltimore, MD 21218 USA
| | - Kari E. Wong
- grid.429438.0Metabolon Inc., Morrisville, NC 27560 USA
| | - James Nicoll
- grid.422945.cZen-Bio, Inc., Research Triangle Park, NC 27709 USA
| | | | - Aubrey D. N. J. de Grey
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA ,SENS Research Foundation, Mountain View, CA 94041 USA
| | | | - Andreas Stahl
- 0000 0001 2181 7878grid.47840.3fUniversity of California, Berkeley, CA 94720 USA
| | - Hal Sternberg
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| |
Collapse
|
37
|
Kurtz A, Elsallab M, Sanzenbacher R, Abou-El-Enein M. Linking Scattered Stem Cell-Based Data to Advance Therapeutic Development. Trends Mol Med 2019; 25:8-19. [DOI: 10.1016/j.molmed.2018.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/20/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023]
|
38
|
Schenk LK, Buchholz B, Henke SF, Michgehl U, Daniel C, Amann K, Kunzelmann K, Pavenstädt H. Nephron-specific knockout of TMEM16A leads to reduced number of glomeruli and albuminuria. Am J Physiol Renal Physiol 2018; 315:F1777-F1786. [PMID: 30156115 DOI: 10.1152/ajprenal.00638.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
TMEM16A is a transmembrane protein from a conserved family of calcium-activated proteins that is highly expressed in the kidney. TMEM16A confers calcium-activated chloride channel activity, which is of importance for various cellular functions in secretory epithelia and involved in secretion-dependent renal cyst growth. However, its specific function in renal physiology has remained elusive so far. Therefore, we generated conditional nephron-specific TMEM16A-knockout mice and found that these animals suffered from albuminuria. Kidney histology demonstrated an intact corticomedullary differentiation and absence of cysts. Electron microscopy showed a normal slit diaphragm. However, the total number of glomeruli and total nephron count was decreased in TMEM16A-knockout animals. At the same time, glomerular diameter was increased, presumably as a result of the hyperfiltration in the remaining glomeruli. TUNEL and PCNA stainings showed increased cell death and increased proliferation. Proximal tubular cilia were intact in young animals, but the number of properly ciliated cells was decreased in older, albuminuric animals. Taken together, our data suggest that TMEM16A may be involved in ureteric bud branching and proper nephron endowment. Loss of TMEM16A resulted in reduced nephron number and, subsequently, albuminuria and tubular damage.
Collapse
Affiliation(s)
- Laura K Schenk
- Internal Medicine D University Hospital of Muenster , Muenster Germany
| | - Bjoern Buchholz
- Department of Nephrology and Hypertension, Friedrich-Alexander University of Erlangen-Nürnberg , Erlangen , Germany
| | - Sebastian F Henke
- Internal Medicine D University Hospital of Muenster , Muenster Germany
| | - Ulf Michgehl
- Internal Medicine D University Hospital of Muenster , Muenster Germany
| | - Christoph Daniel
- Institute for Nephropathology, Friedrich-Alexander University of Erlangen-Nürnberg , Erlangen , Germany
| | - Kerstin Amann
- Institute for Nephropathology, Friedrich-Alexander University of Erlangen-Nürnberg , Erlangen , Germany
| | - Karl Kunzelmann
- Department of Physiology, University of Regensburg , Regensburg Germany
| | | |
Collapse
|
39
|
Courtney JA, Cnota JF, Jones HN. The Role of Abnormal Placentation in Congenital Heart Disease; Cause, Correlate, or Consequence? Front Physiol 2018; 9:1045. [PMID: 30131711 PMCID: PMC6091057 DOI: 10.3389/fphys.2018.01045] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/13/2018] [Indexed: 01/11/2023] Open
Abstract
Congenital heart disease (CHD) is the most common birth defect, affecting ~1% of all live births (van der Linde et al., 2011). Despite improvements in clinical care, it is the leading cause of infant mortality related to birth defects (Yang et al., 2006) and burdens survivors with significant morbidity (Gilboa et al., 2016). Furthermore, CHD accounts for the largest proportion (26.7%) of birth defect-associated hospitalization costs—up to $6.1 billion in 2013 (Arth et al., 2017). Yet after decades of research with a primary focus on genetic etiology, the underlying cause of these defects remains unknown in the majority of cases (Zaidi and Brueckner, 2017). Unexplained CHD may be secondary to undiscovered roles of noncoding genetic, epigenetic, and environmental factors, among others (Russell et al., 2018). Population studies have recently demonstrated that pregnancies complicated by CHD also carry a higher risk of developing pathologies associated with an abnormal placenta including growth disturbances (Puri et al., 2017), preeclampsia (Auger et al., 2015; Brodwall et al., 2016), preterm birth (Laas et al., 2012), and stillbirth (Jorgensen et al., 2014). Both the heart and placenta are vascular organs and develop concurrently; therefore, shared pathways almost certainly direct the development of both. The involvement of placental abnormalities in congenital heart disease, whether causal, commensurate or reactive, is under investigated and given the common developmental window and shared developmental pathways of the heart and placenta and concurrent vasculature development, we propose that further investigation combining clinical data, in vitro, in vivo, and computer modeling is fundamental to our understanding and the potential to develop therapeutics.
Collapse
Affiliation(s)
- Jennifer A Courtney
- Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Division of General Pediatric and Thoracic Surgery, Center for Fetal and Placental Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - James F Cnota
- Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Helen N Jones
- Division of General Pediatric and Thoracic Surgery, Center for Fetal and Placental Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
40
|
Roy AL, Conroy RS. Toward mapping the human body at a cellular resolution. Mol Biol Cell 2018; 29:1779-1785. [PMID: 30058989 PMCID: PMC6085824 DOI: 10.1091/mbc.e18-04-0260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 12/21/2022] Open
Abstract
The adult human body is composed of nearly 37 trillion cells, each with potentially unique molecular characteristics. This Perspective describes some of the challenges and opportunities faced in mapping the molecular characteristics of these cells in specific regions of the body and highlights areas for international collaboration toward the broader goal of comprehensively mapping the human body with cellular resolution.
Collapse
Affiliation(s)
- Ananda L. Roy
- Office of Strategic Coordination, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, MD 20892
| | - Richard S. Conroy
- Office of Strategic Coordination, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
41
|
Tchieu J, Zimmer B, Fattahi F, Amin S, Zeltner N, Chen S, Studer L. A Modular Platform for Differentiation of Human PSCs into All Major Ectodermal Lineages. Cell Stem Cell 2018; 21:399-410.e7. [PMID: 28886367 DOI: 10.1016/j.stem.2017.08.015] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/02/2017] [Accepted: 08/21/2017] [Indexed: 12/22/2022]
Abstract
Directing the fate of human pluripotent stem cells (hPSCs) into different lineages requires variable starting conditions and components with undefined activities, introducing inconsistencies that confound reproducibility and assessment of specific perturbations. Here we introduce a simple, modular protocol for deriving the four main ectodermal lineages from hPSCs. By precisely varying FGF, BMP, WNT, and TGFβ pathway activity in a minimal, chemically defined medium, we show parallel, robust, and reproducible derivation of neuroectoderm, neural crest (NC), cranial placode (CP), and non-neural ectoderm in multiple hPSC lines, on different substrates independently of cell density. We highlight the utility of this system by interrogating the role of TFAP2 transcription factors in ectodermal differentiation, revealing the importance of TFAP2A in NC and CP specification, and performing a small-molecule screen that identified compounds that further enhance CP differentiation. This platform provides a simple stage for systematic derivation of the entire range of ectodermal cell types.
Collapse
Affiliation(s)
- Jason Tchieu
- The Center for Stem Cell Biology, Sloan Kettering Institute, New York, NY 10065, USA; Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Bastian Zimmer
- The Center for Stem Cell Biology, Sloan Kettering Institute, New York, NY 10065, USA; Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Faranak Fattahi
- The Center for Stem Cell Biology, Sloan Kettering Institute, New York, NY 10065, USA; Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA; Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10065, USA
| | - Sadaf Amin
- Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10065, USA; Department of Surgery, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Nadja Zeltner
- The Center for Stem Cell Biology, Sloan Kettering Institute, New York, NY 10065, USA; Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan Kettering Institute, New York, NY 10065, USA; Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA.
| |
Collapse
|
42
|
Singhal A, Agrawal A, Ling J. Regulation of insulin resistance and type II diabetes by hepatitis C virus infection: A driver function of circulating miRNAs. J Cell Mol Med 2018; 22:2071-2085. [PMID: 29411512 PMCID: PMC5867149 DOI: 10.1111/jcmm.13553] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/04/2018] [Indexed: 12/15/2022] Open
Abstract
Hepatitis C virus (HCV) infection is a serious worldwide healthcare issue. Its association with various liver diseases including hepatocellular carcinoma (HCC) is well studied. However, the study on the relationship between HCV infection and the development of insulin resistance and diabetes is very limited. Current research has already elucidated some underlying mechanisms, especially on the regulation of metabolism and insulin signalling by viral proteins. More studies have emerged recently on the correlation between HCV infection‐derived miRNAs and diabetes and insulin resistance. However, no studies have been carried out to directly address if these miRNAs, especially circulating miRNAs, have causal effects on the development of insulin resistance and diabetes. Here, we proposed a new perspective that circulating miRNAs can perform regulatory functions to modulate gene expression in peripheral tissues leading to insulin resistance and diabetes, rather than just a passive factor associated with these pathological processes. The detailed rationales were elaborated through comprehensive literature review and bioinformatic analyses. miR‐122 was identified to be one of the most potential circulating miRNAs to cause insulin resistance. This result along with the idea about the driver function of circulating miRNAs will promote further investigations that eventually lead to the development of novel strategies to treat HCV infection‐associated extrahepatic comorbidities.
Collapse
Affiliation(s)
- Adit Singhal
- Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | | | - Jun Ling
- Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| |
Collapse
|
43
|
Tan L, Wang Z, Li Y. Rabbit models provide insights into bone formation related biological process in atherosclerotic vascular calcification. Biochem Biophys Res Commun 2018; 496:1369-1375. [PMID: 29421657 DOI: 10.1016/j.bbrc.2018.02.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/04/2018] [Indexed: 01/12/2023]
Abstract
Featured as ectopic mineralization along blood vessels, vascular calcification is a major risk factor for a number of prevalent diseases including atherosclerosis. As recent studies identify vascular calcification as a tightly regulated process recapitulating embryonic bone formation, in this study, RNA-seq data generated from rabbit models with inherited or induced hyperlipidemia and atherosclerosis were used, to investigate bone formation related signals and biological processes in atherosclerotic vascular calcification. Evident activation of bone formation was found, together with presence and functioning of bone resorption cell osteoclasts, which were found to possibly also promote bone formation in this disease. Resistance of bone formation and calcification were also found, through down-regulation of pro-ossification regulators and up-regulation of protective inhibitors and Wnt antagonists. Levels of activation and resistance of bone formation differed between the two disease models, suggesting different underlying mechanisms and corresponding treatments. In addition, loss-of-function of protective inhibitors KL and SOST, and possibly crucial role of GPNMB were also highlighted for treatment or further study.
Collapse
Affiliation(s)
- Linuo Tan
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, PR China
| | - Zhen Wang
- Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, PR China.
| | - Yixue Li
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, PR China; Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, PR China; Shanghai Center for Bioinformation Technology, Shanghai Industrial Technology Institute, Shanghai, PR China; Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, PR China.
| |
Collapse
|
44
|
Sundararajan T, Manzardo AM, Butler MG. Functional analysis of schizophrenia genes using GeneAnalytics program and integrated databases. Gene 2018; 641:25-34. [PMID: 29032150 PMCID: PMC6706854 DOI: 10.1016/j.gene.2017.10.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/06/2017] [Accepted: 10/11/2017] [Indexed: 12/14/2022]
Abstract
Schizophrenia (SCZ) is a chronic debilitating neuropsychiatric disorder with multiple risk factors involving numerous complex genetic influences. We examined and updated a master list of clinically relevant and susceptibility genes associated with SCZ reported in the literature and genomic databases dedicated to gene discovery for characterization of SCZ genes. We used the commercially available GeneAnalytics computer-based gene analysis program and integrated genomic databases to create a molecular profile of the updated list of 608 SCZ genes to model their impact in select categories (tissues and cells, diseases, pathways, biological processes, molecular functions, phenotypes and compounds) using specialized GeneAnalytics algorithms. Genes for schizophrenia were predominantly expressed in the cerebellum, cerebral cortex, medulla oblongata, thalamus and hypothalamus. Psychiatric/behavioral disorders incorporating SCZ genes included ADHD, bipolar disorder, autism spectrum disorder and alcohol dependence as well as cancer, Alzheimer's and Parkinson's disease, sleep disturbances and inflammation. Function based analysis of major biological pathways and mechanisms associated with SCZ genes identified glutaminergic receptors (e.g., GRIA1, GRIN2, GRIK4, GRM5), serotonergic receptors (e.g., HTR2A, HTR2C), GABAergic receptors (e.g., GABRA1, GABRB2), dopaminergic receptors (e.g., DRD1, DRD2), calcium-related channels (e.g., CACNA1H, CACNA1B), solute transporters (e.g., SLC1A1, SLC6A2) and for neurodevelopment (e.g., ADCY1, MEF2C, NOTCH2, SHANK3). Biological mechanisms involving synaptic transmission, regulation of membrane potential and transmembrane ion transport were identified as leading molecular functions associated with SCZ genes. Our approach to interrogate SCZ genes and their interactions at various levels has increased our knowledge and insight into the disease process possibly opening new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Tharani Sundararajan
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ann M Manzardo
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS, United States
| | - Merlin G Butler
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS, United States; Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
45
|
Wang Y, Zhao C, Hou Z, Yang Y, Bi Y, Wang H, Zhang Y, Gao S. Unique molecular events during reprogramming of human somatic cells to induced pluripotent stem cells (iPSCs) at naïve state. eLife 2018; 7:29518. [PMID: 29381138 PMCID: PMC5807049 DOI: 10.7554/elife.29518] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 01/29/2018] [Indexed: 12/22/2022] Open
Abstract
Derivation of human naïve cells in the ground state of pluripotency provides promising avenues for developmental biology studies and therapeutic manipulations. However, the molecular mechanisms involved in the establishment and maintenance of human naïve pluripotency remain poorly understood. Using the human inducible reprogramming system together with the 5iLAF naïve induction strategy, integrative analysis of transcriptional and epigenetic dynamics across the transition from human fibroblasts to naïve iPSCs revealed ordered waves of gene network activation sharing signatures with those found during embryonic development from late embryogenesis to pre-implantation stages. More importantly, Transcriptional analysis showed a significant transient reactivation of transcripts with 8-cell-stage-like characteristics in the late stage of reprogramming, suggesting transient activation of gene network with human zygotic genome activation (ZGA)-like signatures during the establishment of naïve pluripotency. Together, Dissecting the naïve reprogramming dynamics by integrative analysis improves the understanding of the molecular features involved in the generation of naïve pluripotency directly from somatic cells.
Collapse
Affiliation(s)
- Yixuan Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chengchen Zhao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhenzhen Hou
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuanyuan Yang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yan Bi
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Hong Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yong Zhang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
46
|
Fu X, He F, Li Y, Shahveranov A, Hutchins AP. Genomic and molecular control of cell type and cell type conversions. CELL REGENERATION 2017; 6:1-7. [PMID: 29348912 PMCID: PMC5769489 DOI: 10.1016/j.cr.2017.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/06/2017] [Accepted: 09/18/2017] [Indexed: 12/17/2022]
Abstract
Organisms are made of a limited number of cell types that combine to form higher order tissues and organs. Cell types have traditionally been defined by their morphologies or biological activity, yet the underlying molecular controls of cell type remain unclear. The onset of single cell technologies, and more recently genomics (particularly single cell genomics), has substantially increased the understanding of the concept of cell type, but has also increased the complexity of this understanding. These new technologies have added a new genome wide molecular dimension to the description of cell type, with genome-wide expression and epigenetic data acting as a cell type ‘fingerprint’ to describe the cell state. Using these genomic fingerprints cell types are being increasingly defined based on specific genomic and molecular criteria, without necessarily a distinct biological function. In this review, we will discuss the molecular definitions of cell types and cell type control, and particularly how endogenous and exogenous transcription factors can control cell types and cell type conversions.
Collapse
|
47
|
Soreq L, Rose J, Soreq E, Hardy J, Trabzuni D, Cookson MR, Smith C, Ryten M, Patani R, Ule J. Major Shifts in Glial Regional Identity Are a Transcriptional Hallmark of Human Brain Aging. Cell Rep 2017; 18:557-570. [PMID: 28076797 PMCID: PMC5263238 DOI: 10.1016/j.celrep.2016.12.011] [Citation(s) in RCA: 269] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 10/04/2016] [Accepted: 12/02/2016] [Indexed: 02/06/2023] Open
Abstract
Gene expression studies suggest that aging of the human brain is determined by a complex interplay of molecular events, although both its region- and cell-type-specific consequences remain poorly understood. Here, we extensively characterized aging-altered gene expression changes across ten human brain regions from 480 individuals ranging in age from 16 to 106 years. We show that astrocyte- and oligodendrocyte-specific genes, but not neuron-specific genes, shift their regional expression patterns upon aging, particularly in the hippocampus and substantia nigra, while the expression of microglia- and endothelial-specific genes increase in all brain regions. In line with these changes, high-resolution immunohistochemistry demonstrated decreased numbers of oligodendrocytes and of neuronal subpopulations in the aging brain cortex. Finally, glial-specific genes predict age with greater precision than neuron-specific genes, thus highlighting the need for greater mechanistic understanding of neuron-glia interactions in aging and late-life diseases.
Collapse
Affiliation(s)
- Lilach Soreq
- Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | | | | - Jamie Rose
- MRC Edinburgh Brain Bank, Academic Neuropathology, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Eyal Soreq
- The Computational, Cognitive and Clinical NeuroImaging Laboratory, Division of Brain Sciences, Imperial College, London SW7 2AZ, UK
| | - John Hardy
- Institute of Neurology, University College London, London WC1N 3BG, UK; Reta Lila Weston Institute of Neurological Studies, UCL ION, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Daniah Trabzuni
- Institute of Neurology, University College London, London WC1N 3BG, UK; Departments of Genetics, King Faisal Specialist Hospital and Research Centre. Riyadh 12713, Saudi Arabia
| | - Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Colin Smith
- MRC Edinburgh Brain Bank, Academic Neuropathology, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Mina Ryten
- Institute of Neurology, University College London, London WC1N 3BG, UK; Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Rickie Patani
- Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Reta Lila Weston Institute of Neurological Studies, UCL ION, 1 Wakefield Street, London WC1N 1PJ, UK; Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh EH8 9YL, UK; Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK.
| | - Jernej Ule
- Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
48
|
Hutchins AP, Yang Z, Li Y, He F, Fu X, Wang X, Li D, Liu K, He J, Wang Y, Chen J, Esteban MA, Pei D. Models of global gene expression define major domains of cell type and tissue identity. Nucleic Acids Res 2017; 45:2354-2367. [PMID: 28426095 PMCID: PMC5389706 DOI: 10.1093/nar/gkx054] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/22/2017] [Indexed: 01/22/2023] Open
Abstract
The current classification of cells in an organism is largely based on their anatomic and developmental origin. Cells types and tissues are traditionally classified into those that arise from the three embryonic germ layers, the ectoderm, mesoderm and endoderm, but this model does not take into account the organization of cell type-specific patterns of gene expression. Here, we present computational models for cell type and tissue specification derived from a collection of 921 RNA-sequencing samples from 272 distinct mouse cell types or tissues. In an unbiased fashion, this analysis accurately predicts the three known germ layers. Unexpectedly, this analysis also suggests that in total there are eight major domains of cell type-specification, corresponding to the neurectoderm, neural crest, surface ectoderm, endoderm, mesoderm, blood mesoderm, germ cells and the embryonic domain. Further, we identify putative genes responsible for specifying the domain and the cell type. This model has implications for understanding trans-lineage differentiation for stem cells, developmental cell biology and regenerative medicine.
Collapse
Affiliation(s)
- Andrew P Hutchins
- Department of Biology, Southern University of Science and Technology of China, Shenzhen, Guangdong 518055, China.,Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Zhongzhou Yang
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Yuhao Li
- Department of Biology, Southern University of Science and Technology of China, Shenzhen, Guangdong 518055, China
| | - Fangfang He
- Department of Biology, Southern University of Science and Technology of China, Shenzhen, Guangdong 518055, China
| | - Xiuling Fu
- Department of Biology, Southern University of Science and Technology of China, Shenzhen, Guangdong 518055, China
| | - Xiaoshan Wang
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Dongwei Li
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Kairong Liu
- Academy of Mathematics and Systems Science, National Center for Mathematics and Interdisciplinary Sciences, Chinese Academy of Sciences, Beijing 100080, China.,Beihang University, Beijing 100191, China
| | - Jiangping He
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Yong Wang
- Academy of Mathematics and Systems Science, National Center for Mathematics and Interdisciplinary Sciences, Chinese Academy of Sciences, Beijing 100080, China
| | - Jiekai Chen
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Miguel A Esteban
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China.,Laboratory of RNA, Chromatin and Human disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Duanqing Pei
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| |
Collapse
|
49
|
Zhu G, Yang H, Chen X, Wu J, Zhang Y, Zhao XM. CSTEA: a webserver for the Cell State Transition Expression Atlas. Nucleic Acids Res 2017; 45:W103-W108. [PMID: 28486666 PMCID: PMC5570201 DOI: 10.1093/nar/gkx402] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/14/2017] [Accepted: 04/28/2017] [Indexed: 01/02/2023] Open
Abstract
Cell state transition is one of the fundamental events in the development of multicellular organisms, and the transition trajectory path has recently attracted much attention. With the accumulation of large amounts of "-omics" data, it is becoming possible to get insights into the molecule mechanisms underlying the transitions between cell states. Here, we present CSTEA (Cell State Transition Expression Atlas), a webserver that organizes, analyzes and visualizes the time-course gene expression data during cell differentiation, cellular reprogramming and trans-differentiation in human and mouse. In particular, CSTEA defines gene signatures for uncharacterized stages during cell state transitions, thereby enabling both experimental and computational biologists to better understand the mechanisms of cell fate determination in mammals. To our best knowledge, CSTEA is the first webserver dedicated to the analysis of time-series gene expression data during cell state transitions. CSTEA is freely available at http://comp-sysbio.org/cstea/.
Collapse
Affiliation(s)
- Guanghui Zhu
- Department of Computer Science and Technology, Tongji University, Shanghai 201804, China
| | - Hui Yang
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Science and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai 200092, China
| | - Xiao Chen
- Department of Computer Science and Technology, Tongji University, Shanghai 201804, China
| | - Jun Wu
- Department of Computer Science and Technology, Tongji University, Shanghai 201804, China
| | - Yong Zhang
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Science and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai 200092, China
| | - Xing-Ming Zhao
- Department of Computer Science and Technology, Tongji University, Shanghai 201804, China
| |
Collapse
|
50
|
Bustamante-Marin XM, Ostrowski LE. Cilia and Mucociliary Clearance. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028241. [PMID: 27864314 DOI: 10.1101/cshperspect.a028241] [Citation(s) in RCA: 384] [Impact Index Per Article: 54.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mucociliary clearance (MCC) is the primary innate defense mechanism of the lung. The functional components are the protective mucous layer, the airway surface liquid layer, and the cilia on the surface of ciliated cells. The cilia are specialized organelles that beat in metachronal waves to propel pathogens and inhaled particles trapped in the mucous layer out of the airways. In health this clearance mechanism is effective, but in patients with primary cilia dyskinesia (PCD) the cilia are abnormal, resulting in deficient MCC and chronic lung disease. This demonstrates the critical importance of the cilia for human health. In this review, we summarize the current knowledge of the components of the MCC apparatus, focusing on the role of cilia in MCC.
Collapse
Affiliation(s)
- Ximena M Bustamante-Marin
- Marsico Lung Institute, Cystic Fibrosis and Pulmonary Diseases Research and Treatment Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Lawrence E Ostrowski
- Marsico Lung Institute, Cystic Fibrosis and Pulmonary Diseases Research and Treatment Center, University of North Carolina, Chapel Hill, North Carolina 27599
| |
Collapse
|