1
|
Guerra‐García A, Balarynová J, Smykal P, von Wettberg EJ, Noble SD, Bett KE. Genetic and transcriptomic analysis of lentil seed imbibition and dormancy in relation to its domestication. THE PLANT GENOME 2025; 18:e70021. [PMID: 40164967 PMCID: PMC11958875 DOI: 10.1002/tpg2.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025]
Abstract
Seed dormancy is an adaptation that delays germination to prevent the start of this process during unsuitable conditions. It is crucial in wild species but its loss was selected during crop domestication to ensure a fast and uniform germination. Water uptake, or imbibition, is the first step of germination. In the Fabaceae family, seeds have physical dormancy, in which seed coats are impermeable to water. We used an interspecific cross between an elite lentil line (Lens culinaris) and a wild lentil (L. orientalis) to investigate the genetic basis of imbibition capacity through quantitative trait locus (QTL) mapping and by using RNA from embryos and seed coats at different development stages, and phenotypic data of seed coat thickness (SCT) and proportion of imbibed seeds (PIS). Both characteristics were consistent throughout different years and locations, suggesting a hereditary component. QTL results suggest that they are each controlled by relatively few loci. Differentially expressed genes (DEGs) within the QTL were considered candidate genes. Two glycosyl-hydrolase genes (a β-glucosidase and a β-galactosidase), which degrade complex polysaccharides in the cell wall, were found among the candidate genes, and one of them had a positive correlation (β-glucosidase) between gene expression and imbibition capacity, and the other gene (β-galactosidase) presented a negative correlation between gene expression and SCT.
Collapse
Affiliation(s)
- Azalea Guerra‐García
- Departamento de Biotecnología y Bioquímica, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV‐IPN)IrapuatoMéxico
| | | | - Petr Smykal
- Department of BotanyPalacký UniversityOlomoucCzech Republic
| | - Eric J von Wettberg
- Department of Agriculture, Landscape, and Environment, Gund Institute for the EnvironmentUniversity of VermontBurlingtonVermontUSA
| | - Scott D. Noble
- Department of Mechanical EngineeringUniversity of SaskatchewanSaskatoonSaskatchewanCanada
| | - Kirstin E. Bett
- Department of Plant SciencesUniversity of SaskatchewanSaskatoonSaskatchewanCanada
| |
Collapse
|
2
|
Ferguson JN, Caproni L, Walter J, Shaw K, Arce-Cubas L, Baines A, Thein MS, Mager S, Taylor G, Cackett L, Mathan J, Vath RL, Martin L, Genty B, Pè ME, Lawson T, Dell’Acqua M, Kromdijk J. A deficient CP24 allele defines variation for dynamic nonphotochemical quenching and photosystem II efficiency in maize. THE PLANT CELL 2025; 37:koaf063. [PMID: 40132112 PMCID: PMC12018801 DOI: 10.1093/plcell/koaf063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 02/12/2025] [Indexed: 03/27/2025]
Abstract
Maize (Zea mays L.) is a global crop species in which CO2 assimilation occurs via the C4 pathway. C4 photosynthesis is typically more efficient than C3 photosynthesis under warm and dry conditions; however, despite this inherent advantage, considerable variation remains in photosynthetic efficiency for C4 species that could be leveraged to benefit crop performance. Here, we investigate the genetic architecture of nonphotochemical quenching (NPQ) and photosystem II (PSII) efficiency using a combination of high-throughput phenotyping and quantitative trait loci (QTL) mapping in a field-grown Multi-parent Advanced Generation Inter-Cross (MAGIC) mapping population. QTL mapping was followed by the identification of putative candidate genes using a combination of genomics, transcriptomics, protein biochemistry, and targeted physiological phenotyping. We identified four genes with a putative causal role in the observed QTL effects. The highest confidence causal gene was found for a large effect QTL for photosynthetic efficiency on chromosome 10, which was underpinned by allelic variation in the expression of the minor PSII antenna protein light harvesting complex photosystem II subunit (LHCB6 or CP24), mainly driven by poor expression associated with the haplotype of the F7 founder line. The historical role of this line in breeding for early flowering time may suggest that the presence of this deficient allele could be enriched in temperate maize germplasm. These findings advance our understanding of the genetic basis of NPQ and PSII efficiency in C4 plants and highlight the potential for breeding strategies aimed at optimizing photosynthetic efficiency in maize.
Collapse
Affiliation(s)
- John N Ferguson
- Department of Plant Sciences, University of Cambridge, Cambridge, Cambridgeshire CB2 3EA, UK
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, UK
| | - Leonardo Caproni
- Institute of Plant Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Julia Walter
- Department of Plant Sciences, University of Cambridge, Cambridge, Cambridgeshire CB2 3EA, UK
| | - Katie Shaw
- Department of Plant Sciences, University of Cambridge, Cambridge, Cambridgeshire CB2 3EA, UK
| | - Lucia Arce-Cubas
- Department of Plant Sciences, University of Cambridge, Cambridge, Cambridgeshire CB2 3EA, UK
| | - Alice Baines
- Department of Plant Sciences, University of Cambridge, Cambridge, Cambridgeshire CB2 3EA, UK
| | - Min Soe Thein
- Institute of Plant Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Svenja Mager
- Institute of Plant Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Georgia Taylor
- Department of Plant Sciences, University of Cambridge, Cambridge, Cambridgeshire CB2 3EA, UK
| | - Lee Cackett
- Department of Plant Sciences, University of Cambridge, Cambridge, Cambridgeshire CB2 3EA, UK
| | - Jyotirmaya Mathan
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, UK
| | - Richard L Vath
- Department of Plant Sciences, University of Cambridge, Cambridge, Cambridgeshire CB2 3EA, UK
| | - Leo Martin
- Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7265, Institut de Biosciences et Biotechnologies Aix-Marseille, Université Aix-Marseille, Saint-Paul-lez-Durance 13108, France
| | - Bernard Genty
- Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7265, Institut de Biosciences et Biotechnologies Aix-Marseille, Université Aix-Marseille, Saint-Paul-lez-Durance 13108, France
| | - Mario Enrico Pè
- Institute of Plant Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Tracy Lawson
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, UK
| | - Matteo Dell’Acqua
- Institute of Plant Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Johannes Kromdijk
- Department of Plant Sciences, University of Cambridge, Cambridge, Cambridgeshire CB2 3EA, UK
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
3
|
Litichevskiy L, Considine M, Gill J, Shandar V, Cox TO, Descamps HC, Wright KM, Amses KR, Dohnalová L, Liou MJ, Tetlak M, Galindo-Fiallos MR, Wong AC, Lundgren P, Kim J, Uhr GT, Rahman RJ, Mason S, Merenstein C, Bushman FD, Raj A, Harding F, Chen Z, Prateek GV, Mullis M, Deighan AG, Robinson L, Tanes C, Bittinger K, Chakraborty M, Bhatt AS, Li H, Barnett I, Davenport ER, Broman KW, Levy M, Cohen RL, Botstein D, Freund A, Di Francesco A, Churchill GA, Li M, Thaiss CA. Gut metagenomes reveal interactions between dietary restriction, ageing and the microbiome in genetically diverse mice. Nat Microbiol 2025:10.1038/s41564-025-01963-3. [PMID: 40164832 DOI: 10.1038/s41564-025-01963-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 02/19/2025] [Indexed: 04/02/2025]
Abstract
The gut microbiome changes with age and has been proposed to mediate the benefit of lifespan-extending interventions such as dietary restriction. However, the causes and consequences of microbiome ageing and the potential of such interventions remain unclear. Here we analysed 2,997 metagenomes collected longitudinally from 913 deeply phenotyped, genetically diverse mice to investigate interactions between the microbiome, ageing, dietary restriction (caloric restriction and fasting), host genetics and a range of health parameters. Among the numerous age-associated microbiome changes that we find in this cohort, increased microbiome uniqueness is the most consistent parameter across a second longitudinal mouse experiment that we performed on inbred mice and a compendium of 4,101 human metagenomes. Furthermore, cohousing experiments show that age-associated microbiome changes may be caused by an accumulation of stochastic environmental exposures (neutral theory) rather than by the influence of an ageing host (selection theory). Unexpectedly, the majority of taxonomic and functional microbiome features show small but significant heritability, and the amount of variation explained by host genetics is similar to ageing and dietary restriction. We also find that more intense dietary interventions lead to larger microbiome changes and that dietary restriction does not rejuvenate the microbiome. Lastly, we find that the microbiome is associated with multiple health parameters, including body composition, immune components and frailty, but not lifespan. Overall, this study sheds light on the factors influencing microbiome ageing and aspects of host physiology modulated by the microbiome.
Collapse
Affiliation(s)
- Lev Litichevskiy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maya Considine
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jasleen Gill
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vasuprada Shandar
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy O Cox
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hélène C Descamps
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Kevin R Amses
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lenka Dohnalová
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan J Liou
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Monika Tetlak
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mario R Galindo-Fiallos
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrea C Wong
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Lundgren
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Junwon Kim
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Giulia T Uhr
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan J Rahman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sydney Mason
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carter Merenstein
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anil Raj
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Fiona Harding
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Zhenghao Chen
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - G V Prateek
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Martin Mullis
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | | | | | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, USA
- Division of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Ami S Bhatt
- Department of Genetics, Stanford University, Stanford, CA, USA
- Divisions of Hematology and Blood & Marrow Transplantation, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Hongzhe Li
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian Barnett
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily R Davenport
- Department of Biology, Pennsylvania State University, University Park, PA, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Karl W Broman
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Maayan Levy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- Arc Institute, Palo Alto, CA, USA
| | | | | | - Adam Freund
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | | | | | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology, Stanford University, Stanford, CA, USA.
- Arc Institute, Palo Alto, CA, USA.
| |
Collapse
|
4
|
Keele GR, Dzieciatkowska M, Hay AM, Vincent M, O'Connor C, Stephenson D, Reisz JA, Nemkov T, Hansen KC, Page GP, Zimring JC, Churchill GA, D'Alessandro A. Genetic architecture of the red blood cell proteome in genetically diverse mice reveals central role of hemoglobin beta cysteine redox status in maintaining circulating glutathione pools. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640676. [PMID: 40093052 PMCID: PMC11908137 DOI: 10.1101/2025.02.27.640676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Red blood cells (RBCs) transport oxygen but accumulate oxidative damage over time, reducing function in vivo and during storage-critical for transfusions. To explore genetic influences on RBC resilience, we profiled proteins, metabolites, and lipids from fresh and stored RBCs obtained from 350 genetically diverse mice. Our analysis identified over 6,000 quantitative trait loci (QTL). Compared to other tissues, prevalence of trans genetic effects over cis reflects the absence of de novo protein synthesis in anucleated RBCs. QTL hotspots at Hbb, Hba, Mon1a, and storage-specific Steap3 linked ferroptosis to hemolysis. Proteasome components clustered at multiple loci, underscoring the importance of degrading oxidized proteins. Post-translational modifications (PTMs) mapped predominantly to hemoglobins, particularly cysteine residues. Loss of reactive C93 in humanized mice (HBB C93A) disrupted redox balance, affecting glutathione pools, protein glutathionylation, and redox PTMs. These findings highlight genetic regulation of RBC oxidation, with implications for transfusion biology and oxidative stress-dependent hemolytic disorders.
Collapse
|
5
|
Biernaskie JM, Garzón-Martínez GA, Corke FMK, Doonan JH. Uncovering the genetic basis of competitiveness and the potential for cooperation in plant groups. Proc Biol Sci 2025; 292:20241984. [PMID: 40068823 PMCID: PMC11896697 DOI: 10.1098/rspb.2024.1984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/19/2024] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Crop productivity was transformed by incorporating dwarfing genes that made plants smaller and less competitive (more cooperative). Beyond such major shifts in plant size, however, it is not clear how much variation in competitiveness remains and how to find its genetic basis. We performed plant density experiments, using 484 lines of the Arabidopsis thaliana multi-parent advanced generation inter-cross population, to compare methods for mapping the genetic basis of plant competitiveness. We first found that a major dwarfing gene, the erecta allele, caused reduced competitiveness and higher group productivity. Then, measuring competitiveness more generally, we found: (i) extensive variation in generic measures of competitiveness that extended beyond the effects of the erecta allele; (ii) a novel genomic region underlying variation in competitiveness; and (iii) that some measures of competitiveness were more useful than others. Our results show how modern genomic resources, including multi-parent populations, could uncover hidden genes for more cooperative crop plants.
Collapse
Affiliation(s)
| | - Gina A. Garzón-Martínez
- Institute of Biological, Environmental, and Rural Sciences, Aberystwyth University, Aberystwyth, Ceredigion, UK
- AGROSAVIA, Mosquera, Colombia
| | - Fiona M. K. Corke
- Institute of Biological, Environmental, and Rural Sciences, Aberystwyth University, Aberystwyth, Ceredigion, UK
| | - John H. Doonan
- Institute of Biological, Environmental, and Rural Sciences, Aberystwyth University, Aberystwyth, Ceredigion, UK
| |
Collapse
|
6
|
D’Alessandro A, Keele GR, Hay A, Nemkov T, Earley EJ, Stephenson D, Vincent M, Deng X, Stone M, Dzieciatkowska M, Hansen KC, Kleinman S, Spitalnik SL, Roubinian N, Norris PJ, Busch MP, Page GP, Stockwell BR, Churchill GA, Zimring JC. Ferroptosis regulates hemolysis in stored murine and human red blood cells. Blood 2025; 145:765-783. [PMID: 39541586 PMCID: PMC11863713 DOI: 10.1182/blood.2024026109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
ABSTRACT Red blood cell (RBC) metabolism regulates hemolysis during aging in vivo and in the blood bank. However, the genetic underpinnings of RBC metabolic heterogeneity and extravascular hemolysis at population scale are incompletely understood. On the basis of the breeding of 8 founder strains with extreme genetic diversity, the Jackson Laboratory diversity outbred population can capture the impact of genetic heterogeneity in like manner to population-based studies. RBCs from 350 outbred mice, either fresh or stored for 7 days, were tested for posttransfusion recovery, as well as metabolomics and lipidomics analyses. Metabolite and lipid quantitative trait loci (QTL) mapped >400 gene-metabolite associations, which we collated into an online interactive portal. Relevant to RBC storage, we identified a QTL hotspot on chromosome 1, mapping on the region coding for the ferrireductase 6-transmembrane epithelial antigen of the prostate 3 (Steap3), a transcriptional target to p53. Steap3 regulated posttransfusion recovery, contributing to a ferroptosis-like process of lipid peroxidation, as validated via genetic manipulation in mice. Translational validation of murine findings in humans, STEAP3 polymorphisms were associated with RBC iron content, lipid peroxidation, and in vitro hemolysis in 13 091 blood donors from the Recipient Epidemiology and Donor Evaluation Study. QTL analyses in humans identified a network of gene products (fatty acid desaturases 1 and 2, epoxide hydrolase 2, lysophosphatidylcholine acetyl-transferase 3, solute carrier family 22 member 16, glucose 6-phosphate dehydrogenase, very long chain fatty acid elongase, and phospholipase A2 group VI) associated with altered levels of oxylipins. These polymorphisms were prevalent in donors of African descent and were linked to allele frequency of hemolysis-linked polymorphisms for Steap3 or p53. These genetic variants were also associated with lower hemoglobin increments in thousands of single-unit transfusion recipients from the vein-to-vein database.
Collapse
Affiliation(s)
- Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, CO
- Omix Technologies Inc, Aurora, CO
| | - Gregory R. Keele
- The Jackson Laboratory, Bar Harbor, ME
- RTI International, Research Triangle Park, NC
| | - Ariel Hay
- Department of Pathology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, CO
- Omix Technologies Inc, Aurora, CO
| | | | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, CO
| | | | - Xutao Deng
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, CO
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, CO
| | - Steven Kleinman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Victoria, BC, Canada
| | | | - Nareg Roubinian
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
- Kaiser Permanente Northern California Division of Research, Oakland, CA
| | - Philip J. Norris
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Michael P. Busch
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | | | - Brent R. Stockwell
- Department of Biological Sciences, Department of Chemistry, and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| | | | - James C. Zimring
- Department of Pathology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
7
|
Reisz JA, Earley EJ, Nemkov T, Key A, Stephenson D, Keele GR, Dzieciatkowska M, Spitalnik SL, Hod EA, Kleinman S, Roubinian NH, Gladwin MT, Hansen KC, Norris PJ, Busch MP, Zimring JC, Churchill GA, Page GP, D'Alessandro A. Arginine metabolism is a biomarker of red blood cell and human aging. Aging Cell 2025; 24:e14388. [PMID: 39478346 PMCID: PMC11822668 DOI: 10.1111/acel.14388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/06/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024] Open
Abstract
Increasing global life expectancy motivates investigations of molecular mechanisms of aging and age-related diseases. This study examines age-associated changes in red blood cells (RBCs), the most numerous host cell in humans. Four cohorts, including healthy individuals and patients with sickle cell disease, were analyzed to define age-dependent changes in RBC metabolism. Over 15,700 specimens from 13,757 humans were examined, a major expansion over previous studies of RBCs in aging. Multi-omics approaches identified chronological age-related alterations in the arginine pathway with increased arginine utilization in RBCs from older individuals. These changes were consistent across healthy and sickle cell disease cohorts and were influenced by genetic variation, sex, and body mass index. Integrating multi-omics data and metabolite quantitative trait loci (mQTL) in humans and 525 diversity outbred mice functionally linked metabolism of arginine during RBC storage to increased vesiculation-a hallmark of RBC aging-and lower post-transfusion hemoglobin increments. Thus, arginine metabolism is a biomarker of RBC and organismal aging, suggesting potential new targets for addressing sequelae of aging.
Collapse
Affiliation(s)
- Julie A. Reisz
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | | | - Travis Nemkov
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Omix Technologies IncAuroraColoradoUSA
| | - Alicia Key
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Daniel Stephenson
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | | | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Steven L. Spitalnik
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Eldad A. Hod
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Steven Kleinman
- University of British ColumbiaVictoriaBritish ColumbiaCanada
| | - Nareg H. Roubinian
- Vitalant Research InstituteSan FranciscoCaliforniaUSA
- Kaiser Permanente Northern California Division of ResearchPleasantonCaliforniaUSA
- Department of Laboratory MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Mark T. Gladwin
- Department of MedicineUniversity of Maryland School of Medicine, University of MarylandBaltimoreMarylandUSA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Omix Technologies IncAuroraColoradoUSA
| | - Philip J. Norris
- Vitalant Research InstituteSan FranciscoCaliforniaUSA
- Department of Laboratory MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Michael P. Busch
- Vitalant Research InstituteSan FranciscoCaliforniaUSA
- Department of Laboratory MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - James C. Zimring
- Department of PathologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | | | | | - Angelo D'Alessandro
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Omix Technologies IncAuroraColoradoUSA
| |
Collapse
|
8
|
Rogers WD, Presley A, Damaj MI, Miles MF. Identification of ethanol analgesia quantitative trait loci and candidate genes in BXD recombinant inbred mouse lines. Addict Biol 2025; 30:e70013. [PMID: 39996569 PMCID: PMC11851315 DOI: 10.1111/adb.70013] [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: 06/28/2024] [Revised: 10/25/2024] [Accepted: 12/07/2024] [Indexed: 02/26/2025]
Abstract
Alcohol consumption produces acute analgesic effects, and people experiencing pain conditions may drink alcohol to alleviate discomfort. However, tolerance to the analgesic properties of alcohol could prompt escalating consumption and dependence. Both nociception and alcohol-induced analgesia are under significant genetic control. Understanding the genetic architecture of these processes could inform better treatment options for people with pain conditions. This study aims to identify quantitative trait loci (QTL) driving variation in ethanol-induced analgesia across BXD recombinant inbred mouse lines. Male and female mice from 62 BXD strains received ethanol or saline oral gavage for five days and were tested for hot plate (HP) latency at baseline, Day 1 and Day 5. QTL mapping of HP phenotypes identified a significant provisional QTL on chromosome 17 for Day 1 HP latency in mice receiving ethanol. An additional highly suggestive QTL was present on chromosome 9 for the difference in pre- and post-ethanol thermal nociception. Candidate genes within QTL support intervals were provisionally identified using HP phenotypic correlations to transcriptomic database, expression QTL analysis and other bioinformatics inquiries. The combined behavioural and bioinformatic analyses yielded strong ethanol analgesia candidate genes, specifically Myo6. Thus, the results of this genetic study of ethanol-induced analgesia in BXD mouse strains may contribute significantly to our understanding of the molecular basis for individual variation in the analgesic response to acute ethanol.
Collapse
Affiliation(s)
- Walker D. Rogers
- Department of Human and Molecular GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- Alcohol Research CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Alyssa Presley
- Department of Pharmacology and ToxicologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - M. Imad Damaj
- Department of Pharmacology and ToxicologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Michael F. Miles
- Department of Human and Molecular GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- Alcohol Research CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of Pharmacology and ToxicologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
9
|
Aydin S, Skelly DA, Dewey H, Mahoney JM, Choi T, Reinholdt LG, Baker CL, Munger SC. Cross cell-type systems genetics reveals the influence of eQTL at multiple points in the developmental trajectory of mouse neural progenitor cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634514. [PMID: 39896448 PMCID: PMC11785210 DOI: 10.1101/2025.01.24.634514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Genetic variation leads to phenotypic variability in pluripotent stem cells that presents challenges for regenerative medicine. Although recent studies have investigated the impact of genetic variation on pluripotency maintenance and differentiation capacity, less is known about how genetic variants affecting the pluripotent state influence gene regulation in later stages of development. Here, we characterized expression of more than 12,000 genes in 127 donor-matched Diversity Outbred (DO) mouse embryonic stem cell (mESC) and neural progenitor cell (mNPC) lines. Quantitative trait locus (QTL) mapping identified 2,947 expression QTL (eQTL) unique to DO mNPCs and 1,113 eQTL observed in both mNPCs and mESCs with highly concordant allele effects. We mapped three eQTL hotspots on Chromosomes (Chrs) 1, 10, and 11 that were unique to mNPCs. Target genes of the Chr 1 hotspot were overrepresented for those involved in mRNA processing, DNA repair, chromatin organization, protein degradation, and cell cycle. Mediation analysis of the Chr 1 hotspot identified Rnf152 as the best candidate mediator expressed in mNPCs, while cross-cell type mediation using mESC gene expression along with partial correlation analysis strongly implicated genetic variant(s) affecting Pign expression in the mESC state as regulating the mNPC Chr 1 eQTL hotspot. Together these findings highlight that many local eQTL confer similar effects on gene expression in multiple cell states; distant eQTL in DO mNPCs are numerous and largely unique to that cell state, with many co-localizing to mNPC-specific hotspots; and mediation analysis across cell types suggests that expression of Pign early in development (mESCs) shapes the transcriptome of the more specialized mNPC state.
Collapse
Affiliation(s)
- Selcan Aydin
- The Jackson Laboratory, Bar Harbor, ME 04609 USA
| | | | - Hannah Dewey
- The Jackson Laboratory, Bar Harbor, ME 04609 USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111 USA
| | | | - Ted Choi
- Predictive Biology, Inc., Carlsbad, CA 92010 USA
| | - Laura G. Reinholdt
- The Jackson Laboratory, Bar Harbor, ME 04609 USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111 USA
| | - Christopher L. Baker
- The Jackson Laboratory, Bar Harbor, ME 04609 USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111 USA
| | - Steven C. Munger
- The Jackson Laboratory, Bar Harbor, ME 04609 USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111 USA
| |
Collapse
|
10
|
Noda Y, Wang F, Chankaew S, Ariga H, Muto C, Iki Y, Ohashi H, Takahashi Y, Sakai H, Iseki K, Ogiso-Tanaka E, Suzui N, Yin YG, Miyoshi Y, Enomoto K, Kawachi N, Somta P, Furukawa J, Tomooka N, Naito K. Diurnal Regulation of SOS Pathway and Sodium Excretion Underlying Salinity Tolerance of Vigna marina. PLANT, CELL & ENVIRONMENT 2025. [PMID: 39853734 DOI: 10.1111/pce.15402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 11/26/2024] [Accepted: 01/10/2025] [Indexed: 01/26/2025]
Abstract
Vigna marina (Barm.) Merr. is adapted to tropical marine beaches and has an outstanding tolerance to salt stress. Given there are growing demands for cultivating crops in saline soil or with saline water, it is important to understand how halophytic species are adapted to the saline environments. Here we revealed by positron-emitting tracer imaging system (PETIS) that V. marina actively excretes sodium from the root during the light period but not the dark period. The following whole genome sequencing accompanied with forward genetic study identified a QTL region harbouring SOS1, encoding plasma membrane Na+/H+ antiporter, which was associated with not only salt tolerance but also the ability of sodium excretion. We also found the QTL region contained a large structural rearrangement that suppressed recombination across ~14 Mbp, fixing multiple gene loci potentially involved in salt tolerance. RNA-seq and promoter analyses revealed SOS1 in V. marina was highly expressed even without salt stress and its promoter shared common cis-regulatory motifs with those exhibiting similar expression profiles. Interestingly, the cis-regulatory motifs seemed installed by a transposable element (TE) insertion. Though not identified by genetic analysis, the transcriptome data also revealed SOS2 transcription was under diurnal regulation, explaining the pattern of sodium excretion together with upregulated expression of SOS1. Altogether, the study elucidated one aspect of the strategy adopted by V. marina to adapt to marine beach, which is highly saline and transpiring.
Collapse
Affiliation(s)
- Yusaku Noda
- Takasaki Institute for Advanced Quantum Science, National Institutes for Quantum Science and Technology (QST), Gunma, Japan
| | - Fanmiao Wang
- Research Center of Genetic Resources, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Sompong Chankaew
- Department of Agronomy, Faculty of Agriculture, Khon Kaen University, Khon Kaen, Thailand
- Department of Agronomy, Faculty of Agriculture at Kamphaeng Saen, Kasetsart University, Nakhon Pathom, Thailand
| | - Hirotaka Ariga
- Research Center of Genetic Resources, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Chiaki Muto
- Research Center of Genetic Resources, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Yurie Iki
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Haruko Ohashi
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yu Takahashi
- Department of Agronomy, Faculty of Agriculture, Khon Kaen University, Khon Kaen, Thailand
| | - Hiroaki Sakai
- Research Center of Advanced Analysis, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Kohtaro Iseki
- Japan International Research Center for Agricultural Sciences, Ibaraki, Japan
| | - Eri Ogiso-Tanaka
- Center for Molecular Biodiversity Research, National Museum of Nature & Science, Ibaraki, Japan
| | - Nobuo Suzui
- Takasaki Institute for Advanced Quantum Science, National Institutes for Quantum Science and Technology (QST), Gunma, Japan
| | - Yong-Gen Yin
- Takasaki Institute for Advanced Quantum Science, National Institutes for Quantum Science and Technology (QST), Gunma, Japan
| | - Yuta Miyoshi
- Takasaki Institute for Advanced Quantum Science, National Institutes for Quantum Science and Technology (QST), Gunma, Japan
| | - Kazuyuki Enomoto
- Takasaki Institute for Advanced Quantum Science, National Institutes for Quantum Science and Technology (QST), Gunma, Japan
| | - Naoki Kawachi
- Takasaki Institute for Advanced Quantum Science, National Institutes for Quantum Science and Technology (QST), Gunma, Japan
| | - Prakit Somta
- Department of Agronomy, Faculty of Agriculture at Kamphaeng Saen, Kasetsart University, Nakhon Pathom, Thailand
| | - Jun Furukawa
- Institute of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan
| | - Norihiko Tomooka
- Research Center of Genetic Resources, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Ken Naito
- Research Center of Genetic Resources, National Agriculture and Food Research Organization, Ibaraki, Japan
| |
Collapse
|
11
|
Atkins K, Garzón-Martínez GA, Lloyd A, Doonan JH, Lu C. Unlocking the power of AI for phenotyping fruit morphology in Arabidopsis. Gigascience 2025; 14:giae123. [PMID: 39937596 PMCID: PMC11816797 DOI: 10.1093/gigascience/giae123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/14/2024] [Accepted: 01/06/2025] [Indexed: 02/14/2025] Open
Abstract
Deep learning can revolutionise high-throughput image-based phenotyping by automating the measurement of complex traits, a task that is often labour-intensive, time-consuming, and prone to human error. However, its precision and adaptability in accurately phenotyping organ-level traits, such as fruit morphology, remain to be fully evaluated. Establishing the links between phenotypic and genotypic variation is essential for uncovering the genetic basis of traits and can also provide an orthologous test of pipeline effectiveness. In this study, we assess the efficacy of deep learning for measuring variation in fruit morphology in Arabidopsis using images from a multiparent advanced generation intercross (MAGIC) mapping family. We trained an instance segmentation model and developed a pipeline to phenotype Arabidopsis fruit morphology, based on the model outputs. Our model achieved strong performance with an average precision of 88.0% for detection and 55.9% for segmentation. Quantitative trait locus analysis of the derived phenotypic metrics of the MAGIC population identified significant loci associated with fruit morphology. This analysis, based on automated phenotyping of 332,194 individual fruits, underscores the capability of deep learning as a robust tool for phenotyping large populations. Our pipeline for quantifying pod morphological traits is scalable and provides high-quality phenotype data, facilitating genetic analysis and gene discovery, as well as advancing crop breeding research.
Collapse
Affiliation(s)
- Kieran Atkins
- National Plant Phenomics Centre, IBERS, Aberystwyth University, Aberystwyth SY23 3EE, UK
| | - Gina A Garzón-Martínez
- Centro de Investigación Tibaitatá, Corporación Colombiana de Investigación Agropecuaria (Agrosavia), Mosquera, Cundinamarca, 250047, Colombia
| | - Andrew Lloyd
- National Plant Phenomics Centre, IBERS, Aberystwyth University, Aberystwyth SY23 3EE, UK
| | - John H Doonan
- National Plant Phenomics Centre, IBERS, Aberystwyth University, Aberystwyth SY23 3EE, UK
| | - Chuan Lu
- Computer Science Department, Aberystwyth University, Aberystwyth SY23 3DB, UK
| |
Collapse
|
12
|
Li Q, Faux P, Wentworth Winchester E, Yang G, Chen Y, Ramírez LM, Fuentes-Guajardo M, Poloni L, Steimetz E, Gonzalez-José R, Acuña V, Bortolini MC, Poletti G, Gallo C, Rothhammer F, Rojas W, Zheng Y, Cox JC, Patel V, Hoffman MP, Ding L, Peng C, Cotney J, Navarro N, Cox TC, Delgado M, Adhikari K, Ruiz-Linares A. PITX2 expression and Neanderthal introgression in HS3ST3A1 contribute to variation in tooth dimensions in modern humans. Curr Biol 2025; 35:131-144.e6. [PMID: 39672157 PMCID: PMC11789201 DOI: 10.1016/j.cub.2024.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/29/2024] [Accepted: 11/15/2024] [Indexed: 12/15/2024]
Abstract
Dental morphology varies greatly throughout evolution, including in the human lineage, but little is known about the biology of this variation. Here, we use multiomics analyses to examine the genetics of variation in tooth crown dimensions. In a human cohort with mixed continental ancestry, we detected genome-wide significant associations at 18 genome regions. One region includes EDAR, a gene known to impact dental features in East Asians. Furthermore, we find that EDAR variants increase the mesiodistal diameter of all teeth, following an anterior-posterior gradient of decreasing strength. Among the 17 novel-associated regions, we replicate 7/13 in an independent human cohort and find that 4/12 orthologous regions affect molar size in mice. Two association signals point to compelling candidate genes. One is ∼61 kb from PITX2, a major determinant of tooth development. Another overlaps HS3ST3A1, a paralogous neighbor of HS3ST3B1, a tooth enamel knot factor. We document the expression of Pitx2 and Hs3st3a1 in enamel knot and dental epithelial cells of developing mouse incisors. Furthermore, associated SNPs in PITX2 and HS3ST3A1 overlap enhancers active in these cells, suggesting a role for these SNPs in gene regulation during dental development. In addition, we document that Pitx2 and Hs3st3a1/Hs3st3b1 knockout mice show alterations in dental morphology. Finally, we find that associated SNPs in HS3ST3A1 are in a DNA tract introgressed from Neanderthals, consistent with an involvement of HS3ST3A1 in tooth size variation during human evolution.
Collapse
Affiliation(s)
- Qing Li
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong District, Shanghai 200433, China; State Key Laboratory of Complex Severe and Rare Diseases, Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing, Dongcheng District, Beijing 100730, China
| | - Pierre Faux
- Aix-Marseille Université, CNRS, EFS, ADES, 27 Boulevard Jean Moulin, Marseille 13005, France; GenPhySE Université de Toulouse, INRAE, ENVT, 24 Chemin de Borde Rouge, 31326 Castanet Tolosan, France
| | - Emma Wentworth Winchester
- Department of Genetics and Genome Sciences, University of Connecticut Health, 400 Farmington Avenue, Farmington, CT 06030, USA
| | - Guangrui Yang
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong District, Shanghai 200433, China; Exchange, Development & Service Center for Science & Technology Talents, Sanlihe Road, Beijing 100045, P.R. China
| | - Yingjie Chen
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong District, Shanghai 200433, China
| | - Luis Miguel Ramírez
- Facultad de Odontología, Universidad de Antioquia, Calle 64 N.º 52-59 Of. 107. Apartado Postal 1226, Medellín, Colombia
| | - Macarena Fuentes-Guajardo
- Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Tarapacá, Avenida 18 de Septiembre 2222, Arica 1000000, Chile
| | - Lauriane Poloni
- Biogéosciences, UMR 6282 CNRS, Université de Bourgogne, Dijon 21000, France; EPHE, PSL University, Paris 75014, France
| | - Emilie Steimetz
- Biogéosciences, UMR 6282 CNRS, Université de Bourgogne, Dijon 21000, France
| | - Rolando Gonzalez-José
- Instituto Patagónico de Ciencias Sociales y Humanas, Centro Nacional Patagónico, CONICET, U9129ACD Puerto Madryn, Argentina
| | - Victor Acuña
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM-Instituto Nacional de Medicina Genómica, México City 4510, México
| | - Maria-Cátira Bortolini
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, 90040-060 Porto Alegre, Brasil
| | - Giovanni Poletti
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, 31 Lima, Perú
| | - Carla Gallo
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, 31 Lima, Perú
| | | | - Winston Rojas
- GENMOL (Genética Molecular), Universidad de Antioquia, 5001000 Medellín, Colombia
| | - Youyi Zheng
- State Key Lab of CAD&CG, Zhejiang University, Yuhangtang Road, Hangzhou 310058, China
| | - James C Cox
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, MO 64108, USA
| | - Vaishali Patel
- Matrix and Morphogenesis Section, NIDCR, NIH, DHHS, Bethesda, MD 20892, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, NIDCR, NIH, DHHS, Bethesda, MD 20892, USA
| | - Li Ding
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong District, Shanghai 200433, China
| | - Chenchen Peng
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong District, Shanghai 200433, China
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut Health, 400 Farmington Avenue, Farmington, CT 06030, USA
| | - Nicolas Navarro
- Biogéosciences, UMR 6282 CNRS, Université de Bourgogne, Dijon 21000, France; EPHE, PSL University, Paris 75014, France
| | - Timothy C Cox
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, MO 64108, USA; Department of Pediatrics, School of Medicine, University of Missouri, 400 N Keene St., Kansas City, MO 64108, USA
| | - Miguel Delgado
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong District, Shanghai 200433, China; División Antropología, Facultad de Ciencias Naturales y Museo, Paseo del Bosque s/n, Universidad Nacional de La Plata, La Plata 1900, República Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Godoy Cruz, 2290 Buenos Aires, República Argentina.
| | - Kaustubh Adhikari
- School of Mathematics and Statistics, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK; Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, Gower Street, London WC1E 6BT, UK.
| | - Andrés Ruiz-Linares
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong District, Shanghai 200433, China; Aix-Marseille Université, CNRS, EFS, ADES, 27 Boulevard Jean Moulin, Marseille 13005, France; Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
13
|
Fu Y, Yuan ZF, Wu L, Peng J, Wang X, High AA. Addressing Sample Mix-Ups: Tools and Approaches for Large-Scale Multi-Omics Studies. Proteomics 2025; 25:e202400271. [PMID: 39659081 DOI: 10.1002/pmic.202400271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024]
Abstract
Advances in high-throughput omics technologies have enabled system-wide characterization of biological samples across multiple molecular levels, such as the genome, transcriptome, and proteome. However, as sample sizes rapidly increase in large-scale multi-omics studies, sample mix-ups have become a prevalent issue, compromising data integrity and leading to erroneous conclusions. The interconnected nature of multi-omics data presents an opportunity to identify and correct these errors. This review examines the potential sources of sample mix-ups and evaluates the methodologies and tools developed for detecting and correcting these errors, with an emphasis on approaches applicable to proteomics data. We categorize existing tools into three main groups: expression/protein quantitative trait loci-based, genotype concordance-based, and gene/protein expression correlation-based approaches. Notably, only a handful of tools currently utilize the proteogenomics approach for correcting sample mix-ups at the proteomics level. Integrating the strengths of current tools across diverse data types could enable the development of more versatile and comprehensive solutions. In conclusion, verifying sample identity is a critical first step to reduce bias and increase precision in subsequent analyses for large-scale multi-omics studies. By leveraging these tools for identifying and correcting sample mix-ups, researchers can significantly improve the reliability and reproducibility of biomedical research.
Collapse
Affiliation(s)
- Yingxue Fu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Zuo-Fei Yuan
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Long Wu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Xusheng Wang
- Department of Neurology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Anthony A High
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
14
|
Reatini B, Pelosi JA, Cang FA, Jiang Q, McKibben MTW, Barker MS, Rieseberg LH, Dlugosch KM. Chromosome-scale Reference Genome and RAD-based Genetic Map of Yellow Starthistle (Centaurea solstitialis) Reveal Putative Structural Variation and QTL Associated With Invader Traits. Genome Biol Evol 2024; 16:evae243. [PMID: 39592405 PMCID: PMC11632367 DOI: 10.1093/gbe/evae243] [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: 06/15/2023] [Revised: 08/19/2024] [Accepted: 09/17/2024] [Indexed: 11/28/2024] Open
Abstract
Invasive species offer outstanding opportunities to identify the genomic sources of variation that contribute to rapid adaptation, as well as the genetic mechanisms facilitating invasions. The Eurasian plant yellow starthistle (Centaurea solstitialis) is highly invasive in North and South American grasslands and known to have evolved increased growth and reproduction during invasion. Here, we develop new genomic resources for C. solstitialis and map the genetic basis of invasiveness traits. We present a chromosome-scale (1N = 8) reference genome using PacBio CLR and Dovetail Omni-C technologies, and functional gene annotation using RNAseq. We find repeat structure typical of the family Asteraceae, with over 25% of gene content derived from ancestral whole-genome duplications (paleologs). Using an F2 mapping population derived from a cross between native and invading parents, with a restriction site-associated DNA (RAD)-based genetic map, we validate the assembly and identify 13 quantitative trait loci underpinning size traits that have evolved during invasion. We find evidence that large effects of quantitative trait loci may be associated with structural variants between native and invading genotypes, including a variant with an overdominant and pleiotropic effect on key invader traits. We also find evidence of significant paleolog enrichment under two quantitative trait loci. Our results add to growing evidence of the importance of structural variants in evolution, and to understanding of the rapid evolution of invaders.
Collapse
Affiliation(s)
- Bryan Reatini
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, USA
| | - Jessie A Pelosi
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, USA
| | - F Alice Cang
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, USA
| | - Qiuyu Jiang
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, USA
| | - Michael T W McKibben
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, USA
| | - Michael S Barker
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, USA
| | - Loren H Rieseberg
- Department of Botany, University of British Columbia, Vancouver, Canada
| | - Katrina M Dlugosch
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, USA
| |
Collapse
|
15
|
Mullis MN, Wright KM, Raj A, Gatti DM, Reifsnyder PC, Flurkey K, Archer JR, Robinson L, Di Francesco A, Svenson KL, Korstanje R, Harrison DE, Ruby JG, Churchill GA. Analysis of lifespan across Diversity Outbred mouse studies identifies multiple longevity-associated loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624531. [PMID: 39605744 PMCID: PMC11601611 DOI: 10.1101/2024.11.20.624531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Lifespan is an integrative phenotype whose genetic architecture is likely to highlight multiple processes with high impact on health and aging. Here, we conduct a genetic meta-analysis of longevity in Diversity Outbred (DO) mice that includes 2,444 animals from three independently conducted lifespan studies. We identify six loci that contribute significantly to lifespan independently of diet and drug treatment, one of which also influences lifespan in a sex-dependent manner, as well as an additional locus with a diet-specific effect on lifespan. Collectively, these loci explain over half of the estimated heritable variation in lifespan across these studies and provide insight into the genetic architecture of lifespan in DO mice.
Collapse
|
16
|
Nelson EA, Tyler AL, Lakusta-Wong T, Lahue KG, Hankes KC, Teuscher C, Lynch RM, Ferris MT, Mahoney JM, Krementsov DN. Analysis of CNS autoimmunity in genetically diverse mice reveals unique phenotypes and mechanisms. JCI Insight 2024; 9:e184138. [PMID: 39325545 PMCID: PMC11601571 DOI: 10.1172/jci.insight.184138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024] Open
Abstract
Multiple sclerosis (MS) is a complex disease with significant heterogeneity in disease course and progression. Genetic studies have identified numerous loci associated with MS risk, but the genetic basis of disease progression remains elusive. To address this, we leveraged the Collaborative Cross (CC), a genetically diverse mouse strain panel, and experimental autoimmune encephalomyelitis (EAE). The 32 CC strains studied captured a wide spectrum of EAE severity, trajectory, and presentation, including severe-progressive, monophasic, relapsing remitting, and axial rotary-EAE (AR-EAE), accompanied by distinct immunopathology. Sex differences in EAE severity were observed in 6 strains. Quantitative trait locus analysis revealed distinct genetic linkage patterns for different EAE phenotypes, including EAE severity and incidence of AR-EAE. Machine learning-based approaches prioritized candidate genes for loci underlying EAE severity (Abcc4 and Gpc6) and AR-EAE (Yap1 and Dync2h1). This work expands the EAE phenotypic repertoire and identifies potentially novel loci controlling unique EAE phenotypes, supporting the hypothesis that heterogeneity in MS disease course is driven by genetic variation.
Collapse
Affiliation(s)
- Emily A. Nelson
- Department of Biomedical and Health Sciences, University of Vermont (UVM), Burlington, Vermont, USA
| | | | | | - Karolyn G. Lahue
- Department of Biomedical and Health Sciences, University of Vermont (UVM), Burlington, Vermont, USA
| | - Katherine C. Hankes
- Department of Biomedical and Health Sciences, University of Vermont (UVM), Burlington, Vermont, USA
| | - Cory Teuscher
- Department of Medicine, UVM, Larner College of Medicine, Burlington, Vermont, USA
| | - Rachel M. Lynch
- Department of Genetics, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
| | - J. Matthew Mahoney
- The Jackson Laboratory, Bar Harbor, Maine, USA
- Department of Neurological Sciences and
| | - Dimitry N. Krementsov
- Department of Biomedical and Health Sciences, University of Vermont (UVM), Burlington, Vermont, USA
| |
Collapse
|
17
|
Morgan AP, Payseur BA. Genetic background affects the strength of crossover interference in house mice. Genetics 2024; 228:iyae146. [PMID: 39241112 PMCID: PMC11538424 DOI: 10.1093/genetics/iyae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/22/2024] [Indexed: 09/08/2024] Open
Abstract
Meiotic recombination is required for faithful chromosome segregation in most sexually reproducing organisms and shapes the distribution of genetic variation in populations. Both the overall rate and the spatial distribution of crossovers vary within and between species. Adjacent crossovers on the same chromosome tend to be spaced more evenly than expected at random, a phenomenon known as crossover interference. Although interference has been observed in many taxa, the factors that influence the strength of interference are not well understood. We used house mice (Mus musculus), a well-established model system for understanding recombination, to study the effects of genetics and age on recombination rate and interference in the male germline. We analyzed crossover positions in 503 progeny from reciprocal F1 hybrids between inbred strains representing the three major subspecies of house mice. Consistent with previous studies, autosomal alleles from M. m. musculus tend to increase recombination rate, while inheriting a M. m. musculus X chromosome decreases recombination rate. Old males transmit an average of 0.6 more crossovers per meiosis (5.0%) than young males, though the effect varies across genetic backgrounds. We show that the strength of crossover interference depends on genotype, providing a rare demonstration that interference evolves over short timescales. Differences between reciprocal F1s suggest that X-linked factors modulate the strength of interference. Our findings motivate additional comparisons of interference among recently diverged species and further examination of the role of paternal age in determining the number and positioning of crossovers.
Collapse
Affiliation(s)
- Andrew P Morgan
- Department of Medicine, University of North Carolina, Campus Box #7085, Chapel Hill, NC 27599-7085, USA
| | - Bret A Payseur
- Laboratory of Genetics, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
18
|
Mignogna KM, Tatom Z, Macleod L, Sergi Z, Nguyen A, Michenkova M, Smith ML, Miles MF. Identification of novel genetic loci and candidate genes for progressive ethanol consumption in diversity outbred mice. Neuropsychopharmacology 2024; 49:1892-1904. [PMID: 38951586 PMCID: PMC11473901 DOI: 10.1038/s41386-024-01902-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/26/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024]
Abstract
Mouse behavioral genetic mapping studies can identify genomic intervals modulating complex traits under well-controlled environmental conditions and have been used to study ethanol behaviors to aid in understanding genetic risk and the neurobiology of alcohol use disorder (AUD). However, historically such studies have produced large confidence intervals, thus complicating identification of potential causal candidate genes. Diversity Outbred (DO) mice offer the ability to perform high-resolution quantitative trait loci (QTL) mapping on a very genetically diverse background, thus facilitating identification of candidate genes. Here, we studied a population of 636 male DO mice with four weeks of intermittent ethanol access via a three-bottle choice procedure, producing a progressive ethanol consumption phenotype. QTL analysis identified 3 significant (Chrs 3, 4, and 12) and 13 suggestive loci for ethanol-drinking behaviors with narrow confidence intervals (1-4 Mbp for significant QTLs). Results suggested that genetic influences on initial versus progressive ethanol consumption were localized to different genomic intervals. A defined set of positional candidate genes were prioritized using haplotype analysis, identified coding polymorphisms, prefrontal cortex transcriptomics data, human GWAS data and prior rodent gene set data for ethanol or other misused substances. These candidates included Car8, the lone gene with a significant cis-eQTL within a Chr 4 QTL for week four ethanol consumption. These results represent the highest-resolution genetic mapping of ethanol consumption behaviors in mice to date, providing identification of novel loci and candidate genes for study in relation to the neurobiology of AUD.
Collapse
Affiliation(s)
- Kristin M Mignogna
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Zachary Tatom
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Lorna Macleod
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Zachary Sergi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Angel Nguyen
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Marie Michenkova
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Maren L Smith
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael F Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA.
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
19
|
Gao AW, El Alam G, Zhu Y, Li W, Sulc J, Li X, Katsyuba E, Li TY, Overmyer KA, Lalou A, Mouchiroud L, Sleiman MB, Cornaglia M, Morel JD, Houtkooper RH, Coon JJ, Auwerx J. High-content phenotypic analysis of a C. elegans recombinant inbred population identifies genetic and molecular regulators of lifespan. Cell Rep 2024; 43:114836. [PMID: 39368088 PMCID: PMC11996002 DOI: 10.1016/j.celrep.2024.114836] [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: 02/08/2024] [Revised: 07/10/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Lifespan is influenced by complex interactions between genetic and environmental factors. Studying those factors in model organisms of a single genetic background limits their translational value for humans. Here, we mapped lifespan determinants in 85 C. elegans recombinant inbred advanced intercross lines (RIAILs). We assessed molecular profiles-transcriptome, proteome, and lipidome-and life-history traits, including lifespan, development, growth dynamics, and reproduction. RIAILs exhibited large variations in lifespan, which correlated positively with developmental time. We validated three longevity modulators, including rict-1, gfm-1, and mltn-1, among the top candidates obtained from multiomics data integration and quantitative trait locus (QTL) mapping. We translated their relevance to humans using UK Biobank data and showed that variants in GFM1 are associated with an elevated risk of age-related heart failure. We organized our dataset as a resource that allows interactive explorations for new longevity targets.
Collapse
Affiliation(s)
- Arwen W Gao
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands.
| | - Gaby El Alam
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Yunyun Zhu
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA
| | - Weisha Li
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Elena Katsyuba
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Nagi Bioscience SA, EPFL Innovation Park, 1025 Saint-Sulpice, Switzerland
| | - Terytty Y Li
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Katherine A Overmyer
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53515, USA
| | - Amelia Lalou
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Laurent Mouchiroud
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Nagi Bioscience SA, EPFL Innovation Park, 1025 Saint-Sulpice, Switzerland
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Matteo Cornaglia
- Nagi Bioscience SA, EPFL Innovation Park, 1025 Saint-Sulpice, Switzerland
| | - Jean-David Morel
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53515, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53506, USA
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
20
|
Di Francesco A, Deighan AG, Litichevskiy L, Chen Z, Luciano A, Robinson L, Garland G, Donato H, Vincent M, Schott W, Wright KM, Raj A, Prateek GV, Mullis M, Hill WG, Zeidel ML, Peters LL, Harding F, Botstein D, Korstanje R, Thaiss CA, Freund A, Churchill GA. Dietary restriction impacts health and lifespan of genetically diverse mice. Nature 2024; 634:684-692. [PMID: 39385029 PMCID: PMC11485257 DOI: 10.1038/s41586-024-08026-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 09/05/2024] [Indexed: 10/11/2024]
Abstract
Caloric restriction extends healthy lifespan in multiple species1. Intermittent fasting, an alternative form of dietary restriction, is potentially more sustainable in humans, but its effectiveness remains largely unexplored2-8. Identifying the most efficacious forms of dietary restriction is key for developing interventions to improve human health and longevity9. Here we performed an extensive assessment of graded levels of caloric restriction (20% and 40%) and intermittent fasting (1 and 2 days fasting per week) on the health and survival of 960 genetically diverse female mice. We show that caloric restriction and intermittent fasting both resulted in lifespan extension in proportion to the degree of restriction. Lifespan was heritable and genetics had a larger influence on lifespan than dietary restriction. The strongest trait associations with lifespan included retention of body weight through periods of handling-an indicator of stress resilience, high lymphocyte proportion, low red blood cell distribution width and high adiposity in late life. Health effects differed between interventions and exhibited inconsistent relationships with lifespan extension. 40% caloric restriction had the strongest lifespan extension effect but led to a loss of lean mass and changes in the immune repertoire that could confer susceptibility to infections. Intermittent fasting did not extend the lifespan of mice with high pre-intervention body weight, and two-day intermittent fasting was associated with disruption of erythroid cell populations. Metabolic responses to dietary restriction, including reduced adiposity and lower fasting glucose, were not associated with increased lifespan, suggesting that dietary restriction does more than just counteract the negative effects of obesity. Our findings indicate that improving health and extending lifespan are not synonymous and raise questions about which end points are the most relevant for evaluating aging interventions in preclinical models and clinical trials.
Collapse
Affiliation(s)
| | | | - Lev Litichevskiy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhenghao Chen
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | | | | | | | | | | | | | - Kevin M Wright
- Calico Life Sciences LLC, South San Francisco, CA, USA
- Actio Biosciences, San Diego, CA, USA
| | - Anil Raj
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - G V Prateek
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Martin Mullis
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Warren G Hill
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Mark L Zeidel
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | - Fiona Harding
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | | | | | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Freund
- Calico Life Sciences LLC, South San Francisco, CA, USA
- Arda Therapeutics, San Carlos, CA, USA
| | | |
Collapse
|
21
|
Tyler AL, Mahoney JM, Keller MP, Baker CN, Gaca M, Srivastava A, Gerdes Gyuricza I, Braun MJ, Rosenthal NA, Attie AD, Churchill GA, Carter GW. Transcripts with high distal heritability mediate genetic effects on complex metabolic traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.613931. [PMID: 39386475 PMCID: PMC11463413 DOI: 10.1101/2024.09.26.613931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Although many genes are subject to local regulation, recent evidence suggests that complex distal regulation may be more important in mediating phenotypic variability. To assess the role of distal gene regulation in complex traits, we combined multi-tissue transcriptomes with physiological outcomes to model diet-induced obesity and metabolic disease in a population of Diversity Outbred mice. Using a novel high-dimensional mediation analysis, we identified a composite transcriptome signature that summarized genetic effects on gene expression and explained 30% of the variation across all metabolic traits. The signature was heritable, interpretable in biological terms, and predicted obesity status from gene expression in an independently derived mouse cohort and multiple human studies. Transcripts contributing most strongly to this composite mediator frequently had complex, distal regulation distributed throughout the genome. These results suggest that trait-relevant variation in transcription is largely distally regulated, but is nonetheless identifiable, interpretable, and translatable across species.
Collapse
|
22
|
Desaint H, Héreil A, Belinchon-Moreno J, Carretero Y, Pelpoir E, Pascal M, Brault M, Dumont D, Lecompte F, Laugier P, Duboscq R, Bitton F, Grumic M, Giraud C, Ferrante P, Giuliano G, Sunseri F, Causse M. Integration of QTL and transcriptome approaches for the identification of genes involved in tomato response to nitrogen deficiency. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:5880-5896. [PMID: 38869971 DOI: 10.1093/jxb/erae265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/12/2024] [Indexed: 06/15/2024]
Abstract
Optimizing plant nitrogen (N) usage and inhibiting N leaching loss in the soil-crop system is crucial to maintaining crop yield and reducing environmental pollution. This study aimed at identifying quantitative trait loci (QTLs) and differentially expressed genes (DEGs) between two N treatments in order to list candidate genes related to nitrogen-related contrasting traits in tomato varieties. We characterized a genetic diversity core-collection (CC) and a multi-parental advanced generation intercross (MAGIC) tomato population grown in a greenhouse under two nitrogen levels and assessed several N-related traits and mapped QTLs. Transcriptome response under the two N conditions was also investigated through RNA sequencing of fruit and leaves in four parents of the MAGIC population. Significant differences in response to N input reduction were observed at the phenotypic level for biomass and N-related traits. Twenty-seven QTLs were detected for three target traits (leaf N content, leaf nitrogen balance index, and petiole NO3- content), 10 and six in the low and high N condition, respectively, while 19 QTLs were identified for plasticity traits. At the transcriptome level, 4752 and 2405 DEGs were detected between the two N conditions in leaves and fruits, respectively, among which 3628 (50.6%) in leaves and 1717 (71.4%) in fruit were genotype specific. When considering all the genotypes, 1677 DEGs were shared between organs or tissues. Finally, we integrated DEG and QTL analyses to identify the most promising candidate genes. The results highlighted a complex genetic architecture of N homeostasis in tomato and novel putative genes useful for breeding tomato varieties requiring less N input.
Collapse
Affiliation(s)
| | | | | | | | | | - Michel Pascal
- INRAE, UR407, Pathologie Végétale, 84143 Montfavet, France
| | | | | | | | | | | | | | | | | | - Paola Ferrante
- Italian National Agency for New technologies, Energy and Sustainable Economic Development (ENEA), Casaccia Res Ctr, Via Anguillarese 301, 00123 Rome, Italy
| | - Giovanni Giuliano
- Italian National Agency for New technologies, Energy and Sustainable Economic Development (ENEA), Casaccia Res Ctr, Via Anguillarese 301, 00123 Rome, Italy
| | | | | |
Collapse
|
23
|
Nemkov T, Stephenson D, Earley EJ, Keele GR, Hay A, Key A, Haiman ZB, Erickson C, Dzieciatkowska M, Reisz JA, Moore A, Stone M, Deng X, Kleinman S, Spitalnik SL, Hod EA, Hudson KE, Hansen KC, Palsson BO, Churchill GA, Roubinian N, Norris PJ, Busch MP, Zimring JC, Page GP, D'Alessandro A. Biological and genetic determinants of glycolysis: Phosphofructokinase isoforms boost energy status of stored red blood cells and transfusion outcomes. Cell Metab 2024; 36:1979-1997.e13. [PMID: 38964323 PMCID: PMC11374506 DOI: 10.1016/j.cmet.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/04/2024] [Accepted: 06/07/2024] [Indexed: 07/06/2024]
Abstract
Mature red blood cells (RBCs) lack mitochondria and thus exclusively rely on glycolysis to generate adenosine triphosphate (ATP) during aging in vivo or storage in blood banks. Here, we leveraged 13,029 volunteers from the Recipient Epidemiology and Donor Evaluation Study to identify associations between end-of-storage levels of glycolytic metabolites and donor age, sex, and ancestry-specific genetic polymorphisms in regions encoding phosphofructokinase 1, platelet (detected in mature RBCs); hexokinase 1 (HK1); and ADP-ribosyl cyclase 1 and 2 (CD38/BST1). Gene-metabolite associations were validated in fresh and stored RBCs from 525 Diversity Outbred mice and via multi-omics characterization of 1,929 samples from 643 human RBC units during storage. ATP and hypoxanthine (HYPX) levels-and the genetic traits linked to them-were associated with hemolysis in vitro and in vivo, both in healthy autologous transfusion recipients and in 5,816 critically ill patients receiving heterologous transfusions, suggesting their potential as markers to improve transfusion outcomes.
Collapse
Affiliation(s)
- Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA; Omix Technologies Inc., Aurora, CO, USA
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Ariel Hay
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Alicia Key
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
| | - Zachary B Haiman
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Christopher Erickson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
| | | | - Mars Stone
- Vitalant Research Institute, San Francisco, CA, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Xutao Deng
- Vitalant Research Institute, San Francisco, CA, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Steven L Spitalnik
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Eldad A Hod
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Krystalyn E Hudson
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA; Omix Technologies Inc., Aurora, CO, USA
| | - Bernhard O Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | - Nareg Roubinian
- Vitalant Research Institute, San Francisco, CA, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA; Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
| | - Philip J Norris
- Vitalant Research Institute, San Francisco, CA, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Michael P Busch
- Vitalant Research Institute, San Francisco, CA, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - James C Zimring
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | | | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA; Omix Technologies Inc., Aurora, CO, USA.
| |
Collapse
|
24
|
Poirion OB, Zuo W, Spruce C, Baker CN, Daigle SL, Olson A, Skelly DA, Chesler EJ, Baker CL, White BS. Enhlink infers distal and context-specific enhancer-promoter linkages. Genome Biol 2024; 25:235. [PMID: 39223609 PMCID: PMC11368035 DOI: 10.1186/s13059-024-03374-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Enhlink is a computational tool for scATAC-seq data analysis, facilitating precise interrogation of enhancer function at the single-cell level. It employs an ensemble approach incorporating technical and biological covariates to infer condition-specific regulatory DNA linkages. Enhlink can integrate multi-omic data for enhanced specificity, when available. Evaluation with simulated and real data, including multi-omic datasets from the mouse striatum and novel promoter capture Hi-C data, demonstrate that Enhlink outperfoms alternative methods. Coupled with eQTL analysis, it identified a putative super-enhancer in striatal neurons. Overall, Enhlink offers accuracy, power, and potential for revealing novel biological insights in gene regulation.
Collapse
Affiliation(s)
| | - Wulin Zuo
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | | | | | - Ashley Olson
- The Jackson Laboratory, Bar Harbor, ME, USA
- Center for Systems Neurogenetics of Addiction at The Jackson Laboratory, Bar Harbor, ME, USA
| | | | - Elissa J Chesler
- The Jackson Laboratory, Bar Harbor, ME, USA
- Center for Systems Neurogenetics of Addiction at The Jackson Laboratory, Bar Harbor, ME, USA
| | - Christopher L Baker
- The Jackson Laboratory, Bar Harbor, ME, USA
- Center for Systems Neurogenetics of Addiction at The Jackson Laboratory, Bar Harbor, ME, USA
| | - Brian S White
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| |
Collapse
|
25
|
Koch RL, Stanton JB, McClatchy S, Churchill GA, Craig SW, Williams DN, Johns ME, Chase KR, Thiesfeldt DL, Flynt JC, Pazdro R. Discovery of genomic loci for liver health and steatosis reveals overlap with glutathione redox genetics. Redox Biol 2024; 75:103248. [PMID: 38917671 PMCID: PMC11254179 DOI: 10.1016/j.redox.2024.103248] [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: 03/08/2024] [Revised: 05/27/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver condition in the United States, encompassing a wide spectrum of liver pathologies including steatosis, steatohepatitis, fibrosis, and cirrhosis. Despite its high prevalence, there are no medications currently approved by the Food and Drug Administration for the treatment of NAFLD. Recent work has suggested that NAFLD has a strong genetic component and identifying causative genes will improve our understanding of the molecular mechanisms contributing to NAFLD and yield targets for future therapeutic investigations. Oxidative stress is known to play an important role in NAFLD pathogenesis, yet the underlying mechanisms accounting for disturbances in redox status are not entirely understood. To better understand the relationship between the glutathione redox system and signs of NAFLD in a genetically-diverse population, we measured liver weight, serum biomarkers aspartate aminotransferase (AST) and alanine aminotransferase (ALT), and graded liver pathology in a large cohort of Diversity Outbred mice. We compared hepatic endpoints to those of the glutathione redox system previously measured in the livers and kidneys of the same mice, and we screened for statistical and genetic associations using the R/qtl2 software. We discovered several novel genetic loci associated with markers of liver health, including loci that were associated with both liver steatosis and glutathione redox status. Candidate genes within each locus point to possible new mechanisms underlying the complex relationship between NAFLD and the glutathione redox system, which could have translational implications for future studies targeting NAFLD pathology.
Collapse
Affiliation(s)
- Rebecca L Koch
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - James B Stanton
- Department of Pathology, University of Georgia, Athens, GA, USA, 30602
| | | | | | - Steven W Craig
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Darian N Williams
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Mallory E Johns
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Kylah R Chase
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Dana L Thiesfeldt
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Jessica C Flynt
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Robert Pazdro
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602.
| |
Collapse
|
26
|
van Megen WH, de Baaij JHF, Churchill GA, Devuyst O, Hoenderop JGJ, Korstanje R. Genetic drivers of age-related changes in urinary magnesium excretion. Physiol Genomics 2024; 56:634-647. [PMID: 39037434 PMCID: PMC11460537 DOI: 10.1152/physiolgenomics.00119.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/23/2024] Open
Abstract
Although age-dependent alterations in urinary magnesium (Mg2+) excretion have been described, the underlying mechanism remains elusive. As heritability significantly contributes to variations in urinary Mg2+ excretion, we measured urinary Mg2+ excretion at different ages in a cohort of genetically variable Diversity Outbred (DO) mice. Compared with animals aged 6 mo, an increase in Mg2+ excretion was observed at 12 and 18 mo. Quantitative trait locus (QTL) analysis revealed an association of a locus on chromosome 10 with Mg2+ excretion at 6 mo of age, with Oit3 (encoding oncoprotein-induced transcript 3; OIT3) as our primary candidate gene. To study the possible role of OIT3 in renal Mg2+ handling, we generated and characterized Oit3 knockout (Oit3-/-) mice. Although a slightly lower serum Mg2+ concentration was present in male Oit3-/- mice, this effect was not observed in female Oit3-/- mice. In addition, urinary Mg2+ excretion and the expression of renal magnesiotropic genes were unaltered in Oit3-/- mice. For animals aged 12 and 18 mo, QTL analysis revealed an association with a locus on chromosome 19, which contains the gene encoding TRPM6, a known Mg2+ channel involved in renal Mg2+ reabsorption. Comparison with RNA sequencing (RNA-Seq) data revealed that Trpm6 mRNA expression is inversely correlated with the QTL effect, implying that TRPM6 may be involved in age-dependent changes in urinary Mg2+ excretion in mice. In conclusion, we show here that variants in Oit3 and Trpm6 are associated with urinary Mg2+ excretion at distinct periods of life, although OIT3 is unlikely to affect renal Mg2+ handling.NEW & NOTEWORTHY Aging increased urinary magnesium (Mg2+) excretion in mice. We show here that variation in Oit3, a candidate gene for the locus associated with Mg2+ excretion in young mice, is unlikely to be involved as knockout of Oit3 did not affect Mg2+ excretion. Differences in the expression of the renal Mg2+ channel TRPM6 may contribute to the variation in urinary Mg2+ excretion in older mice.
Collapse
Affiliation(s)
- Wouter H van Megen
- Department of Medical Biosciences, Radboudumc, Nijmegen, The Netherlands
| | | | | | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Ron Korstanje
- The Jackson Laboratory, Bar Harbor, Maine, United States
| |
Collapse
|
27
|
Ueda Y, Kondo K, Saito H, Pariasca-Tanaka J, Takanashi H, Ranaivo HN, Rakotondramanana M, Wissuwa M. Characterization of quantitative trait loci from DJ123 ( aus) independently affecting panicle structure traits in indica rice cultivar IR64. MOLECULAR BREEDING : NEW STRATEGIES IN PLANT IMPROVEMENT 2024; 44:57. [PMID: 39228865 PMCID: PMC11366739 DOI: 10.1007/s11032-024-01494-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
The rice panicle is the principal organ to influence productivity and traits affecting panicle architecture determine sink size and yield potential. Improving panicle architecture may be effective in increasing yield under low-input conditions, but which traits are of importance under such conditions and how they are genetically controlled is not well understood. Using recombinant inbred lines (RILs) derived from a cross between a modern variety IR64 and a low fertility tolerant accession DJ123, quantitative trait locus (QTL) mapping was conducted under high soil fertility in Japan and low fertility in Madagascar. Among QTL for panicle length (PL) detected, the DJ123 allele increased rachis length at qCL1 and qPL9, while the IR64 allele increased primary branch length at qPL7. DJ123 further contributed two QTL for grain width whereas IR64 contributed two grain length QTL. Analysis of lines carrying different combinations of detected QTL indicates that rachis and primary branch lengths are independently regulated, explaining strong transgressive segregation for PL. The positive effects of PL-related QTL were further confirmed by a genome-wide analysis of allelic states in two breeding lines that had been selected repeatedly for total panicle weight per plant under low input conditions. This study provides the genetic basis for complex panicle architecture in rice and will aid in designing an ideal panicle architecture that leads to increased yield under low fertility conditions. Supplementary information The online version contains supplementary material available at 10.1007/s11032-024-01494-5.
Collapse
Affiliation(s)
- Yoshiaki Ueda
- Crop, Livestock and Environment Division, Japan International Research Center for Agricultural Sciences, Tsukuba, Ibaraki Japan
| | - Katsuhiko Kondo
- Crop, Livestock and Environment Division, Japan International Research Center for Agricultural Sciences, Tsukuba, Ibaraki Japan
- Present Address: Research Institute of Rice Production and Technology Co., Ltd., Toyoake, Aichi Japan
| | - Hiroki Saito
- Tropical Agriculture Research Front, Japan International Research Center for Agricultural Sciences, Ishigaki, Okinawa Japan
| | - Juan Pariasca-Tanaka
- Crop, Livestock and Environment Division, Japan International Research Center for Agricultural Sciences, Tsukuba, Ibaraki Japan
| | - Hideki Takanashi
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Harisoa Nicole Ranaivo
- Rice Research Department, The National Center for Applied Research On Rural Development (FOFIFA), Antananarivo, Madagascar
| | - Mbolatantely Rakotondramanana
- Rice Research Department, The National Center for Applied Research On Rural Development (FOFIFA), Antananarivo, Madagascar
| | - Matthias Wissuwa
- Crop, Livestock and Environment Division, Japan International Research Center for Agricultural Sciences, Tsukuba, Ibaraki Japan
- PhenoRob Cluster & Institute of Crop Science and Resource Conservation (INRES), University of Bonn, Bonn, Germany
| |
Collapse
|
28
|
Chowdhury M, Johansen M, Davison A. Continuous variation in the shell colour of the snail Cepaea nemoralis is associated with the colour locus of the supergene. J Evol Biol 2024; 37:1091-1100. [PMID: 39066609 DOI: 10.1093/jeb/voae093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/11/2024] [Accepted: 07/25/2024] [Indexed: 07/28/2024]
Abstract
While the shell of the land snail Cepaea nemoralis is typically classed as yellow, pink, or brown, the reality is that colour variation is continuously distributed. To further understand the origin of the continuous variation, we used crosses of C. nemoralis to compare quantitative measures of the colour with the inferred genotype of the underlying supergene locus. We also used a recently developed linkage map to find quantitative trait loci that may influence colour. The results show that the colour locus of the supergene-at around 31.385 cM on linkage group 11-is involved in determining the quantitative chromatic differences that are perceptible to human vision. We also found some evidence that variation within colour classes may be due to allelic variation at or around the supergene. There are likely other unlinked loci involved in determining colour within classes, but confirmation will require greater statistical power. Although not investigated here, environmental factors, including diet, may also impact upon variation within colour types.
Collapse
Affiliation(s)
- Mehrab Chowdhury
- School of Life Sciences, University Park, University of Nottingham, Nottingham, United Kingdom
| | - Margrethe Johansen
- School of Life Sciences, University Park, University of Nottingham, Nottingham, United Kingdom
| | - Angus Davison
- School of Life Sciences, University Park, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
29
|
Yi X, Kemppainen P, Reid K, Chen Y, Rastas P, Fraimout A, Merilä J. Heterogeneous genomic architecture of skeletal armour traits in sticklebacks. J Evol Biol 2024; 37:995-1008. [PMID: 39073424 DOI: 10.1093/jeb/voae083] [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: 02/01/2024] [Revised: 04/28/2024] [Accepted: 07/27/2024] [Indexed: 07/30/2024]
Abstract
Whether populations adapt to similar selection pressures using the same underlying genetic variants depends on population history and the distribution of standing genetic variation at the metapopulation level. Studies of sticklebacks provide a case in point: when colonizing and adapting to freshwater habitats, three-spined sticklebacks (Gasterosteus aculeatus) with high gene flow tend to fix the same adaptive alleles in the same major loci, whereas nine-spined sticklebacks (Pungitius pungitius) with limited gene flow tend to utilize a more heterogeneous set of loci. In accordance with this, we report results of quantitative trait locus (QTL) analyses using a backcross design showing that lateral plate number variation in the western European nine-spined sticklebacks mapped to 3 moderate-effect QTL, contrary to the major-effect QTL in three-spined sticklebacks and different from the 4 QTL previously identified in the eastern European nine-spined sticklebacks. Furthermore, several QTL were identified associated with variation in lateral plate size, and 3 moderate-effect QTL with body size. Together, these findings indicate more heterogenous and polygenic genetic underpinnings of skeletal armour variation in nine-spined than three-spined sticklebacks, indicating limited genetic parallelism underlying armour trait evolution in the family Gasterostidae.
Collapse
Affiliation(s)
- Xueling Yi
- Area of Ecology and Biodiversity, School of Biological Sciences, University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Petri Kemppainen
- Area of Ecology and Biodiversity, School of Biological Sciences, University of Hong Kong, Hong Kong, Hong Kong SAR
- Ecological Genetics Research Unit, Organismal and Evolutionary Biology Programme, University of Helsinki, Helsinki, Finland
| | - Kerry Reid
- Area of Ecology and Biodiversity, School of Biological Sciences, University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Ying Chen
- Area of Ecology and Biodiversity, School of Biological Sciences, University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Pasi Rastas
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Antoine Fraimout
- Ecological Genetics Research Unit, Organismal and Evolutionary Biology Programme, University of Helsinki, Helsinki, Finland
| | - Juha Merilä
- Area of Ecology and Biodiversity, School of Biological Sciences, University of Hong Kong, Hong Kong, Hong Kong SAR
- Ecological Genetics Research Unit, Organismal and Evolutionary Biology Programme, University of Helsinki, Helsinki, Finland
| |
Collapse
|
30
|
Morgan AP, Payseur BA. Genetic background affects the strength of crossover interference in house mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596233. [PMID: 38854148 PMCID: PMC11160618 DOI: 10.1101/2024.05.28.596233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Meiotic recombination is required for faithful chromosome segregation in most sexually reproducing organisms and shapes the distribution of genetic variation in populations. Both the overall rate and the spatial distribution of crossovers vary within and between species. Adjacent crossovers on the same chromosome tend to be spaced more evenly than expected at random, a phenomenon known as crossover interference. Although interference has been observed in many taxa, the factors that influence the strength of interference are not well understood. We used house mice (Mus musculus), a well-established model system for understanding recombination, to study the effects of genetics and age on recombination rate and interference in the male germline. We analyzed crossover positions in 503 progeny from reciprocal F1 hybrids between inbred strains representing the three major subspecies of house mice. Consistent with previous studies, autosomal alleles from M. m. musculus tend to increase recombination rate, while inheriting a M. m. musculus X chromosome decreases recombination rate. Old males transmit an average of 0.6 more crossovers per meiosis (5.0%) than young males, though the effect varies across genetic backgrounds. We show that the strength of crossover interference depends on genotype, providing a rare demonstration that interference evolves over short timescales. Differences between reciprocal F1s suggest that X-linked factors modulate the strength of interference. Our findings motivate additional comparisons of interference among recently diverged species and further examination of the role of paternal age in determining the number and positioning of crossovers.
Collapse
Affiliation(s)
- Andrew P Morgan
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Bret A Payseur
- Laboratory of Genetics, University of Wisconsin, Madison, WI
| |
Collapse
|
31
|
Nagarajan A, Scoggin K, Adams LG, Threadgill D, Andrews-Polymenis H. Identification of a genetic region linked to tolerance to MRSA infection using Collaborative Cross mice. PLoS Genet 2024; 20:e1011378. [PMID: 39178306 PMCID: PMC11407622 DOI: 10.1371/journal.pgen.1011378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 09/17/2024] [Accepted: 07/29/2024] [Indexed: 08/25/2024] Open
Abstract
Staphylococcus aureus (S. aureus) colonizes humans asymptomatically but can also cause opportunistic infections, ranging from mild skin infections to severe life-threatening conditions. Resistance and tolerance are two ways a host can survive an infection. Resistance is limiting the pathogen burden, while tolerance is limiting the health impact of a given pathogen burden. In previous work, we established that collaborative cross (CC) mouse line CC061 is highly susceptible to Methicillin-resistant S. aureus infection (MRSA, USA300), while CC024 is tolerant. To identify host genes involved in tolerance after S. aureus infection, we crossed CC061 mice and CC024 mice to generate F1 and F2 populations. Survival after MRSA infection in the F1 and F2 generations was 65% and 55% and followed a complex dominant inheritance pattern for the CC024 increased survival phenotype. Colonization in F2 animals was more extreme than in their parents, suggesting successful segregation of genetic factors. We identified a Quantitative Trait Locus (QTL) peak on chromosome 7 for survival and weight change after infection. In this QTL, the WSB/EiJ (WSB) allele was present in CC024 mice and contributed to their MRSA tolerant phenotype. Two genes, C5ar1 and C5ar2, have high-impact variants in this region. C5ar1 and C5ar2 are receptors for the complement factor C5a, an anaphylatoxin that can trigger a massive immune response by binding to these receptors. We hypothesize that C5a may have altered binding to variant receptors in CC024 mice, reducing damage caused by the cytokine storm and resulting in the ability to tolerate a higher pathogen burden and longer survival.
Collapse
Affiliation(s)
- Aravindh Nagarajan
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - Kristin Scoggin
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - L Garry Adams
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - David Threadgill
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, United States of America
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas, United States of America
- Department of Biochemistry & Biophysics and Department of Nutrition, Texas A&M University, College Station, Texas, United States of America
| | - Helene Andrews-Polymenis
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
32
|
Orteu A, Hornett EA, Reynolds LA, Warren IA, Hurst GDD, Martin SH, Jiggins CD. Optix and cortex/ivory/mir-193 again: the repeated use of two mimicry hotspot loci. Proc Biol Sci 2024; 291:20240627. [PMID: 39045691 PMCID: PMC11267468 DOI: 10.1098/rspb.2024.0627] [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: 03/15/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
The extent to which evolution is repeatable has been a debated topic among evolutionary biologists. Although rewinding the tape of life perhaps would not lead to the same outcome every time, repeated evolution of analogous genes for similar functions has been extensively reported. Wing phenotypes of butterflies and moths have provided a wealth of examples of gene re-use, with certain 'hotspot loci' controlling wing patterns across diverse taxa. Here, we present an example of convergent evolution in the molecular genetic basis of Batesian wing mimicry in two Hypolimnas butterfly species. We show that mimicry is controlled by variation near cortex/ivory/mir-193, a known butterfly hotspot locus. By dissecting the genetic architecture of mimicry in Hypolimnas misippus and Hypolimnas bolina, we present evidence that distinct non-coding regions control the development of white pattern elements in the forewing and hindwing of the two species, suggesting independent evolution, and that no structural variation is found at the locus. Finally, we also show that orange coloration in H. bolina is associated with optix, a well-known patterning gene. Overall, our study once again implicates variation near the hotspot loci cortex/ivory/mir-193 and optix in butterfly wing mimicry and thereby highlights the repeatability of adaptive evolution.
Collapse
Affiliation(s)
- Anna Orteu
- Tree of Life Programme, Wellcome Sanger Institute, Hinxton, UK
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Emily A. Hornett
- Institute of Infection, Veterinary and Ecological Science, University of Liverpool, Liverpool, UK
- Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Department of Biology, University of Oxford, Oxford, UK
| | - Louise A. Reynolds
- Institute of Infection, Veterinary and Ecological Science, University of Liverpool, Liverpool, UK
| | - Ian A. Warren
- Tree of Life Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Gregory D. D. Hurst
- Institute of Infection, Veterinary and Ecological Science, University of Liverpool, Liverpool, UK
| | - Simon H. Martin
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
33
|
Pierotti S, Welz B, Osuna-López M, Fitzgerald T, Wittbrodt J, Birney E. Genotype imputation in F2 crosses of inbred lines. BIOINFORMATICS ADVANCES 2024; 4:vbae107. [PMID: 39077633 PMCID: PMC11286293 DOI: 10.1093/bioadv/vbae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/04/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Motivation Crosses among inbred lines are a fundamental tool for the discovery of genetic loci associated with phenotypes of interest. In organisms for which large reference panels or SNP chips are not available, imputation from low-pass whole-genome sequencing is an effective method for obtaining genotype data from a large number of individuals. To date, a structured analysis of the conditions required for optimal genotype imputation has not been performed. Results We report a systematic exploration of the effect of several design variables on imputation performance in F2 crosses of inbred medaka lines using the imputation software STITCH. We determined that, depending on the number of samples, imputation performance reaches a plateau when increasing the per-sample sequencing coverage. We also systematically explored the trade-offs between cost, imputation accuracy, and sample numbers. We developed a computational pipeline to streamline the process, enabling other researchers to perform a similar cost-benefit analysis on their population of interest. Availability and implementation The source code for the pipeline is available at https://github.com/birneylab/stitchimpute. While our pipeline has been developed and tested for an F2 population, the software can also be used to analyse populations with a different structure.
Collapse
Affiliation(s)
- Saul Pierotti
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Hinxton, Cambridge CB101SD, United Kingdom
| | - Bettina Welz
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg 69120, Germany
| | - Mireia Osuna-López
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg 69117, Germany
| | - Tomas Fitzgerald
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Hinxton, Cambridge CB101SD, United Kingdom
| | - Joachim Wittbrodt
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg 69120, Germany
| | - Ewan Birney
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Hinxton, Cambridge CB101SD, United Kingdom
| |
Collapse
|
34
|
Skelly DA, Graham JP, Cheng M, Furuta M, Walter A, Stoklasek TA, Yang H, Stearns TM, Poirion O, Zhang JG, Grassmann JDS, Luo D, Flynn WF, Courtois ET, Chang CH, Serreze DV, Menghi F, Reinholdt LG, Liu ET. Mapping the genetic landscape establishing a tumor immune microenvironment favorable for anti-PD-1 response in mice and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603136. [PMID: 39071392 PMCID: PMC11275897 DOI: 10.1101/2024.07.11.603136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Identifying host genetic factors modulating immune checkpoint inhibitor (ICI) efficacy has been experimentally challenging because of variations in both host and tumor genomes, differences in the microbiome, and patient life exposures. Utilizing the Collaborative Cross (CC) multi-parent mouse genetic resource population, we developed an approach that fixes the tumor genomic configuration while varying host genetics. With this approach, we discovered that response to anti-PD-1 (aPD1) immunotherapy was significantly heritable in four distinct murine tumor models (H2 between 0.18-0.40). For the MC38 colorectal carcinoma system (H2 = 0.40), we mapped four significant ICI response quantitative trait loci (QTL) localized to mouse chromosomes (mChr) 5, 9, 15 and 17, and identified significant epistatic interactions between specific QTL pairs. Differentially expressed genes within these QTL were highly enriched for immune genes and pathways mediating allograft rejection and graft vs host disease. Using a cross species analytical approach, we found a core network of 48 genes within the four QTLs that showed significant prognostic value for overall survival in aPD1 treated human cohorts that outperformed all other existing validated immunotherapy biomarkers, especially in human tumors of the previously defined immune subtype 4. Functional blockade of two top candidate immune targets within the 48 gene network, GM-CSF and high affinity IL-2/IL-15 signaling, completely abrogated the MC38 tumor transcriptional response to aPD1 therapy in vivo. Thus, we have established a powerful cross species in vivo platform capable of uncovering host genetic factors that establish the tumor immune microenvironment configuration propitious for ICI response.
Collapse
Affiliation(s)
- Daniel A. Skelly
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - John P. Graham
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | | | - Mayuko Furuta
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Andrew Walter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | | | | | | | - Olivier Poirion
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Ji-Gang Zhang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | | | - Diane Luo
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - William F. Flynn
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Elise T. Courtois
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- OB/Gyn Department, UConn Health, Farmington, CT, USA
| | - Chih-Hao Chang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - David V. Serreze
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - Francesca Menghi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Edison T. Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| |
Collapse
|
35
|
Cui L, Sun M, Zhang L, Zhu H, Kong Q, Dong L, Liu X, Zeng X, Sun Y, Zhang H, Duan L, Li W, Zou C, Zhang Z, Cai W, Ming Y, Lübberstedt T, Liu H, Yang X, Li X. Quantitative trait locus analysis of gray leaf spot resistance in the maize IBM Syn10 DH population. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2024; 137:183. [PMID: 39002016 DOI: 10.1007/s00122-024-04694-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
KEY MESSAGE The exploration and dissection of a set of QTLs and candidate genes for gray leaf spot disease resistance using two fully assembled parental genomes may help expedite maize resistance breeding. The fungal disease of maize known as gray leaf spot (GLS), caused by Cercospora zeae-maydis and Cercospora zeina, is a significant concern in China, Southern Africa, and the USA. Resistance to GLS is governed by multiple genes with an additive effect and is influenced by both genotype and environment. The most effective way to reduce the cost of production is to develop resistant hybrids. In this study, we utilized the IBM Syn 10 Doubled Haploid (IBM Syn10 DH) population to identify quantitative trait loci (QTLs) associated with resistance to gray leaf spot (GLS) in multiple locations. Analysis of seven distinct environments revealed a total of 58 QTLs, 49 of which formed 12 discrete clusters distributed across chromosomes 1, 2, 3, 4, 8 and 10. By comparing these findings with published research, we identified colocalized QTLs or GWAS loci within eleven clustering intervals. By integrating transcriptome data with genomic structural variations between parental individuals, we identified a total of 110 genes that exhibit both robust disparities in gene expression and structural alterations. Further analysis revealed 19 potential candidate genes encoding conserved resistance gene domains, including putative leucine-rich repeat receptors, NLP transcription factors, fucosyltransferases, and putative xyloglucan galactosyltransferases. Our results provide a valuable resource and linked loci for GLS marker resistance selection breeding in maize.
Collapse
Affiliation(s)
- Lina Cui
- Institute of Plant Protection, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, China
- Key Laboratory of Integrated Pest Management on Crops in Southwest China, Ministry of Agriculture, Chengdu, 610066, China
| | - Mingfei Sun
- State Key Laboratory of Wheat Improvement, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Lin Zhang
- Department of Agronomy, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Hongjie Zhu
- State Key Laboratory of Wheat Improvement, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Qianqian Kong
- School of Agriculture, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Ling Dong
- Department of Agronomy, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Xianjun Liu
- Department of Agronomy, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Xing Zeng
- Department of Agronomy, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Yanjie Sun
- Suihua Branch, Heilongjiang Academy of Agricultural Sciences, Suihua, 152052, China
| | - Haiyan Zhang
- Institute of Plant Protection, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, China
- Key Laboratory of Integrated Pest Management on Crops in Southwest China, Ministry of Agriculture, Chengdu, 610066, China
| | - Luyao Duan
- Institute of Plant Protection, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, China
- Key Laboratory of Integrated Pest Management on Crops in Southwest China, Ministry of Agriculture, Chengdu, 610066, China
| | - Wenyi Li
- Institute of Plant Protection, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, China
- Key Laboratory of Integrated Pest Management on Crops in Southwest China, Ministry of Agriculture, Chengdu, 610066, China
| | - Chengjia Zou
- Institute of Plant Protection, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, China
- Key Laboratory of Integrated Pest Management on Crops in Southwest China, Ministry of Agriculture, Chengdu, 610066, China
| | - Zhenyu Zhang
- Institute of Plant Protection, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, China
- Key Laboratory of Integrated Pest Management on Crops in Southwest China, Ministry of Agriculture, Chengdu, 610066, China
| | - WeiLi Cai
- Institute of Plant Protection, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, China
- Key Laboratory of Integrated Pest Management on Crops in Southwest China, Ministry of Agriculture, Chengdu, 610066, China
| | - Yulin Ming
- Liangshan Seed Management Station, Xichang, 615000, China
| | | | - Hongjun Liu
- State Key Laboratory of Wheat Improvement, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Xuerong Yang
- State Key Laboratory of Wheat Improvement, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China.
| | - Xiao Li
- Institute of Plant Protection, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, China.
- Key Laboratory of Integrated Pest Management on Crops in Southwest China, Ministry of Agriculture, Chengdu, 610066, China.
| |
Collapse
|
36
|
St John ME, Dunker JC, Richards EJ, Romero S, Martin CH. Parallel evolution of integrated craniofacial traits in trophic specialist pupfishes. Ecol Evol 2024; 14:e11640. [PMID: 38979003 PMCID: PMC11228360 DOI: 10.1002/ece3.11640] [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: 01/24/2024] [Revised: 05/14/2024] [Accepted: 06/13/2024] [Indexed: 07/10/2024] Open
Abstract
Populations may adapt to similar environments via parallel or non-parallel genetic changes, but the frequency of these alternative mechanisms and underlying contributing factors are still poorly understood outside model systems. We used QTL mapping to investigate the genetic basis of highly divergent craniofacial traits between the scale-eater (Cyprinodon desquamator) and molluscivore (C. brontotheroides) pupfish adapting to two different hypersaline lake environments on San Salvador Island, Bahamas. We lab-reared F2 scale-eater x molluscivore intercrosses from two different lake populations, estimated linkage maps, scanned for significant QTL for 29 skeletal and craniofacial traits, female mate preference, and sex. We compared the location of QTL between lakes to quantify parallel and non-parallel genetic changes. We detected significant QTL for six craniofacial traits in at least one lake. However, nearly all shared QTL loci were associated with a different craniofacial trait within each lake. Therefore, our estimate of parallel evolution of craniofacial genetic architecture could range from one out of six identical trait QTL (low parallelism) to five out of six integrated trait QTL (high parallelism). We suggest that pleiotropy and trait integration can affect estimates of parallel evolution, particularly within rapid radiations. We also observed increased adaptive introgression in shared QTL regions, suggesting that gene flow contributed to parallel evolution. Overall, our results suggest that the same genomic regions may contribute to parallel adaptation across integrated suites of craniofacial traits, rather than specific traits, and highlight the need for a more expansive definition of parallel evolution.
Collapse
Affiliation(s)
| | - Julia C Dunker
- Department of Integrative Biology University of California Berkeley California USA
| | - Emilie J Richards
- Department of Ecology, Evolution and Behavior University of Minnesota Minneapolis Minnesota USA
| | - Stephanie Romero
- Department of Evolution and Ecology University of California Davis California USA
| | - Christopher H Martin
- Department of Integrative Biology University of California Berkeley California USA
- Museum of Vertebrate Zoology University of California Berkeley California USA
| |
Collapse
|
37
|
Parikh C, Glenn RA, Shi Y, Chatterjee K, Swanzey EE, Singer S, Do SC, Zhan Y, Furuta Y, Tahiliani M, Apostolou E, Polyzos A, Koche R, Mezey JG, Vierbuchen T, Stadtfeld M. Genetic variation modulates susceptibility to aberrant DNA hypomethylation and imprint deregulation in naïve pluripotent stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600805. [PMID: 38979237 PMCID: PMC11230387 DOI: 10.1101/2024.06.26.600805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Naïve pluripotent stem cells (nPSC) frequently undergo pathological and not readily reversible loss of DNA methylation marks at imprinted gene loci. This abnormality poses a hurdle for using pluripotent cell lines in biomedical applications and underscores the need to identify the causes of imprint instability in these cells. We show that nPSCs from inbred mouse strains exhibit pronounced strain-specific susceptibility to locus-specific deregulation of imprinting marks during reprogramming to pluripotency and upon culture with MAP kinase inhibitors, a common approach to maintain naïve pluripotency. Analysis of genetically highly diverse nPSCs from the Diversity Outbred (DO) stock confirms that genetic variation is a major determinant of epigenome stability in pluripotent cells. We leverage the variable DNA hypomethylation in DO lines to identify several trans-acting quantitative trait loci (QTLs) that determine epigenome stability at either specific target loci or genome-wide. Candidate factors encoded by two multi-target QTLs on chromosomes 4 and 17 suggest specific transcriptional regulators that contribute to DNA methylation maintenance in nPSCs. We propose that genetic variants represent candidate biomarkers to identify pluripotent cell lines with desirable properties and might serve as entry points for the targeted engineering of nPSCs with stable epigenomes. Highlights Naïve pluripotent stem cells from distinct inbred mouse strains exhibit variable DNA methylation levels at imprinted gene loci.The vulnerability of pluripotent stem cells to loss of genomic imprinting caused by MAP kinase inhibition strongly differs between inbred mouse strains.Genetically diverse pluripotent stem cell lines from Diversity Outbred mouse stock allow the identification of quantitative trait loci controlling DNA methylation stability.Genetic variants may serve as biomarkers to identify naïve pluripotent stem cell lines that are epigenetically stable in specific culture conditions.
Collapse
|
38
|
Oliveira TY, Merkenschlager J, Eisenreich T, Bortolatto J, Yao KH, Gatti DM, Churchill GA, Nussenzweig MC, Breton G. Quantitative trait loci mapping provides insights into the genetic regulation of dendritic cell numbers in mouse tissues. Cell Rep 2024; 43:114296. [PMID: 38823019 PMCID: PMC11726347 DOI: 10.1016/j.celrep.2024.114296] [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: 12/07/2023] [Revised: 04/02/2024] [Accepted: 05/14/2024] [Indexed: 06/03/2024] Open
Abstract
To explore the influence of genetics on homeostatic regulation of dendritic cell (DC) numbers, we present a screen of DCs and their progenitors in lymphoid and non-lymphoid tissues in Collaborative Cross (CC) and Diversity Outbred (DO) mice. We report 30 and 71 loci with logarithm of the odds (LOD) scores >8.18 and ranging from 6.67 to 8.19, respectively. The analysis reveals the highly polygenic and pleiotropic architecture of this complex trait, including many of the previously identified genetic regulators of DC development and maturation. Two SNPs in genes potentially underlying variation in DC homeostasis, a splice variant in Gramd4 (rs235532740) and a missense variant in Orai3 (rs216659754), are confirmed by gene editing using CRISPR-Cas9. Gramd4 is a central regulator of DC homeostasis that impacts the entire DC lineage, and Orai3 regulates cDC2 numbers in tissues. Overall, the data reveal a large number of candidate genes regulating DC homeostasis in vivo.
Collapse
Affiliation(s)
- Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Julia Merkenschlager
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Thomas Eisenreich
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Juliana Bortolatto
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Kai-Hui Yao
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | | | | | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute (HHMI), The Rockefeller University, New York, NY 10065, USA.
| | - Gaëlle Breton
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
39
|
Li X, Morel JD, Sulc J, De Masi A, Lalou A, Benegiamo G, Poisson J, Liu Y, Von Alvensleben GVG, Gao AW, Bou Sleiman M, Auwerx J. Systems genetics of metabolic health in the BXD mouse genetic reference population. Cell Syst 2024; 15:497-509.e3. [PMID: 38866010 DOI: 10.1016/j.cels.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/29/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024]
Abstract
Susceptibility to metabolic syndrome (MetS) is dependent on genetics, environment, and gene-by-environment interactions, rendering the study of underlying mechanisms challenging. The majority of experiments in model organisms do not incorporate genetic variation and lack specific evaluation criteria for MetS. Here, we derived a continuous metric, the metabolic health score (MHS), based on standard clinical parameters and defined its molecular signatures in the liver and circulation. In human UK Biobank, the MHS associated with MetS status and was predictive of future disease incidence, even in individuals without MetS. Using quantitative trait locus analyses in mice, we found two MHS-associated genetic loci and replicated them in unrelated mouse populations. Through a prioritization scheme in mice and human genetic data, we identified TNKS and MCPH1 as candidates mediating differences in the MHS. Our findings provide insights into the molecular mechanisms sustaining metabolic health across species and uncover likely regulators. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Xiaoxu Li
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jean-David Morel
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Alessia De Masi
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Amélia Lalou
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Giorgia Benegiamo
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Johanne Poisson
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Yasmine Liu
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Giacomo V G Von Alvensleben
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Arwen W Gao
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
40
|
Rogers WD, White A, Damaj MI, Miles MF. Identification of ethanol analgesia quantitative trait loci and candidate genes in BXD recombinant inbred mouse lines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599372. [PMID: 38948869 PMCID: PMC11212936 DOI: 10.1101/2024.06.17.599372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Alcohol consumption produces acute analgesic effects, and people experiencing pain conditions may drink alcohol to alleviate discomfort. However, tolerance to the analgesic properties of alcohol could prompt escalating consumption and dependence. Both nociception and alcohol-induced analgesia are under significant genetic control. Understanding the genetic architecture of these processes could inform better treatment options for people with pain conditions. This study aims to identify quantitative trait loci (QTL) driving variation in ethanol-induced analgesia across BXD recombinant inbred mouse lines. Male and female mice from 62 BXD strains received ethanol or saline oral gavage for five days and were tested for hot plate (HP) latency at baseline, Day 1, and Day 5. QTL mapping of HP phenotypes identified a significant provisional QTL on chromosome 17 for Day 1 HP latency in mice receiving ethanol. An additional highly suggestive QTL was present on chromosome 9 for the difference in pre- and post-ethanol thermal nociception. Candidate genes within QTL support intervals were provisionally identified using HP phenotypic correlations to transcriptomic database, expression QTL analysis, and other bioinformatics inquiries. The combined behavioral and bioinformatic analyses yielded strong ethanol analgesia candidate genes, specifically Myo6. Thus, the results of this genetic study of ethanol-induced analgesia in BXD mouse strains may contribute significantly to our understanding of the molecular basis for individual variation in the analgesic response to acute ethanol.
Collapse
Affiliation(s)
- Walker D. Rogers
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States
- Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Alyssa White
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Michael F. Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States
- Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States
| |
Collapse
|
41
|
Nemkov T, Key A, Stephenson D, Earley EJ, Keele GR, Hay A, Amireault P, Casimir M, Dussiot M, Dzieciatkowska M, Reisz JA, Deng X, Stone M, Kleinman S, Spitalnik SL, Hansen KC, Norris PJ, Churchill GA, Busch MP, Roubinian N, Page GP, Zimring JC, Arduini A, D’Alessandro A. Genetic regulation of carnitine metabolism controls lipid damage repair and aging RBC hemolysis in vivo and in vitro. Blood 2024; 143:2517-2533. [PMID: 38513237 PMCID: PMC11208298 DOI: 10.1182/blood.2024023983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/22/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
ABSTRACT Recent large-scale multiomics studies suggest that genetic factors influence the chemical individuality of donated blood. To examine this concept, we performed metabolomics analyses of 643 blood units from volunteers who donated units of packed red blood cells (RBCs) on 2 separate occasions. These analyses identified carnitine metabolism as the most reproducible pathway across multiple donations from the same donor. We also measured l-carnitine and acyl-carnitines in 13 091 packed RBC units from donors in the Recipient Epidemiology and Donor Evaluation study. Genome-wide association studies against 879 000 polymorphisms identified critical genetic factors contributing to interdonor heterogeneity in end-of-storage carnitine levels, including common nonsynonymous polymorphisms in genes encoding carnitine transporters (SLC22A16, SLC22A5, and SLC16A9); carnitine synthesis (FLVCR1 and MTDH) and metabolism (CPT1A, CPT2, CRAT, and ACSS2), and carnitine-dependent repair of lipids oxidized by ALOX5. Significant associations between genetic polymorphisms on SLC22 transporters and carnitine pools in stored RBCs were validated in 525 Diversity Outbred mice. Donors carrying 2 alleles of the rs12210538 SLC22A16 single-nucleotide polymorphism exhibited the lowest l-carnitine levels, significant elevations of in vitro hemolysis, and the highest degree of vesiculation, accompanied by increases in lipid peroxidation markers. Separation of RBCs by age, via in vivo biotinylation in mice, and Percoll density gradients of human RBCs, showed age-dependent depletions of l-carnitine and acyl-carnitine pools, accompanied by progressive failure of the reacylation process after chemically induced membrane lipid damage. Supplementation of stored murine RBCs with l-carnitine boosted posttransfusion recovery, suggesting this could represent a viable strategy to improve RBC storage quality.
Collapse
Affiliation(s)
- Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
- Omix Technologies Inc, Aurora, CO
| | - Alicia Key
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Eric J. Earley
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC
| | - Gregory R. Keele
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC
- The Jackson Laboratory, Bar Harbor, ME
| | - Ariel Hay
- Department of Pathology, University of Virginia, Charlottesville, VA
| | - Pascal Amireault
- Université Paris Cité et Université des Antilles, INSERM, Biologie Intégrée du Globule Rouge, Paris, France
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
| | - Madeleine Casimir
- Université Paris Cité et Université des Antilles, INSERM, Biologie Intégrée du Globule Rouge, Paris, France
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
| | - Michaël Dussiot
- Université Paris Cité et Université des Antilles, INSERM, Biologie Intégrée du Globule Rouge, Paris, France
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Xutao Deng
- Vitalant Research Institute, San Francisco, CA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Steve Kleinman
- The University of British Columbia, Victoria, BC, Canada
| | | | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Philip J. Norris
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | | | - Michael P. Busch
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Nareg Roubinian
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
- Kaiser Permanente Northern California Division of Research, Oakland, CA
| | - Grier P. Page
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC
| | - James C. Zimring
- Department of Pathology, University of Virginia, Charlottesville, VA
| | - Arduino Arduini
- Department of Research and Development, CoreQuest Sagl, Lugano, Switzerland
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
- Omix Technologies Inc, Aurora, CO
| |
Collapse
|
42
|
Nemkov T, Stephenson D, Earley EJ, Keele GR, Hay A, Key A, Haiman Z, Erickson C, Dzieciatkowska M, Reisz JA, Moore A, Stone M, Deng X, Kleinman S, Spitalnik SL, Hod EA, Hudson KE, Hansen KC, Palsson BO, Churchill GA, Roubinian N, Norris PJ, Busch MP, Zimring JC, Page GP, D'Alessandro A. Biological and Genetic Determinants of Glycolysis: Phosphofructokinase Isoforms Boost Energy Status of Stored Red Blood Cells and Transfusion Outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.11.557250. [PMID: 38260479 PMCID: PMC10802247 DOI: 10.1101/2023.09.11.557250] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Mature red blood cells (RBCs) lack mitochondria, and thus exclusively rely on glycolysis to generate adenosine triphosphate (ATP) during aging in vivo or storage in the blood bank. Here we leveraged 13,029 volunteers from the Recipient Epidemiology and Donor Evaluation Study to identify an association between end-of-storage levels of glycolytic metabolites and donor age, sex, and ancestry-specific genetic polymorphisms in regions encoding phosphofructokinase 1, platelet (detected in mature RBCs), hexokinase 1, ADP-ribosyl cyclase 1 and 2 (CD38/BST1). Gene-metabolite associations were validated in fresh and stored RBCs from 525 Diversity Outbred mice, and via multi-omics characterization of 1,929 samples from 643 human RBC units during storage. ATP and hypoxanthine levels - and the genetic traits linked to them - were associated with hemolysis in vitro and in vivo, both in healthy autologous transfusion recipients and in 5,816 critically ill patients receiving heterologous transfusions, suggesting their potential as markers to improve transfusion outcomes. eTOC and Highlights Highlights Blood donor age and sex affect glycolysis in stored RBCs from 13,029 volunteers;Ancestry, genetic polymorphisms in PFKP, HK1, CD38/BST1 influence RBC glycolysis;Modeled PFKP effects relate to preventing loss of the total AXP pool in stored RBCs;ATP and hypoxanthine are biomarkers of hemolysis in vitro and in vivo.
Collapse
|
43
|
D'Alessandro A, Keele GR, Hay A, Nemkov T, Earley EJ, Stephenson D, Vincent M, Deng X, Stone M, Dzieciatkowska M, Hansen KC, Kleinman S, Spitalnik SL, Roubinian NH, Norris PJ, Busch MP, Page GP, Stockwell BR, Churchill GA, Zimring JC. Ferroptosis regulates hemolysis in stored murine and human red blood cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598512. [PMID: 38915523 PMCID: PMC11195277 DOI: 10.1101/2024.06.11.598512] [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
Red blood cell (RBC) metabolism regulates hemolysis during aging in vivo and in the blood bank. Here, we leveraged a diversity outbred mouse population to map the genetic drivers of fresh/stored RBC metabolism and extravascular hemolysis upon storage and transfusion in 350 mice. We identify the ferrireductase Steap3 as a critical regulator of a ferroptosis-like process of lipid peroxidation. Steap3 polymorphisms were associated with RBC iron content, in vitro hemolysis, and in vivo extravascular hemolysis both in mice and 13,091 blood donors from the Recipient Epidemiology and Donor evaluation Study. Using metabolite Quantitative Trait Loci analyses, we identified a network of gene products (FADS1/2, EPHX2 and LPCAT3) - enriched in donors of African descent - associated with oxylipin metabolism in stored human RBCs and related to Steap3 or its transcriptional regulator, the tumor protein TP53. Genetic variants were associated with lower in vivo hemolysis in thousands of single-unit transfusion recipients. Highlights Steap3 regulates lipid peroxidation and extravascular hemolysis in 350 diversity outbred miceSteap3 SNPs are linked to RBC iron, hemolysis, vesiculation in 13,091 blood donorsmQTL analyses of oxylipins identified ferroptosis-related gene products FADS1/2, EPHX2, LPCAT3Ferroptosis markers are linked to hemoglobin increments in transfusion recipients. Graphical abstract
Collapse
|
44
|
Zhang L, Su B, Huang J, Zhang L, Chang Y, Hu G. Fine Mapping of QTLs for Alkaline Tolerance in Crucian Carp ( Carassius auratus) Using Genome-Wide SNP Markers. Genes (Basel) 2024; 15:751. [PMID: 38927687 PMCID: PMC11202869 DOI: 10.3390/genes15060751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Crucian carp (Carassius auratus) is widely distributed in the world and has become an economically freshwater fish. The population in Lake Dali Nur can tolerate the extreme alkaline environment with alkalinity over 50 mmol/L (pH 9.6), thus providing a special model for exploring alkali-tolerant molecular markers in an extremely alkaline environment. In this study, we constructed a high-density and high-resolution linkage map with 16,224 SNP markers based on genotyping-by-sequencing (GBS) consisting of 152 progenies and conducted QTL studies for alkali-tolerant traits. The total length of the linkage map was 3918.893 cM, with an average distance of 0.241 cM. Two QTLs for the ammonia-N-tolerant trait were detected on LG27 and LG45. A QTL for the urea-N-tolerant trait was detected on LG27. Interestingly, mapping the two QTLs on LG27 revealed that the mapped genes were both located in the intron of CDC42. GO functional annotation and KEGG enrichment analysis results indicated that the biological functions might be involved in the cell cycle, cellular senescence, MAPK, and Ras signaling pathways. These findings suggest that CDC42 may play an important role in the process of dealing with extremely alkaline environments.
Collapse
Affiliation(s)
- Liang Zhang
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin 150070, China;
| | - Baofeng Su
- Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats Heilongjiang Province, Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin 150070, China; (B.S.); (J.H.); (L.Z.)
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Jing Huang
- Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats Heilongjiang Province, Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin 150070, China; (B.S.); (J.H.); (L.Z.)
| | - Limin Zhang
- Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats Heilongjiang Province, Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin 150070, China; (B.S.); (J.H.); (L.Z.)
| | - Yumei Chang
- Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats Heilongjiang Province, Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin 150070, China; (B.S.); (J.H.); (L.Z.)
| | - Guo Hu
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin 150070, China;
| |
Collapse
|
45
|
Glenn RA, Do SC, Guruvayurappan K, Corrigan EK, Santini L, Medina-Cano D, Singer S, Cho H, Liu J, Broman K, Czechanski A, Reinholdt L, Koche R, Furuta Y, Kunz M, Vierbuchen T. A PLURIPOTENT STEM CELL PLATFORM FOR IN VITRO SYSTEMS GENETICS STUDIES OF MOUSE DEVELOPMENT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597758. [PMID: 38895226 PMCID: PMC11185710 DOI: 10.1101/2024.06.06.597758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The directed differentiation of pluripotent stem cells (PSCs) from panels of genetically diverse individuals is emerging as a powerful experimental system for characterizing the impact of natural genetic variation on developing cell types and tissues. Here, we establish new PSC lines and experimental approaches for modeling embryonic development in a genetically diverse, outbred mouse stock (Diversity Outbred mice). We show that a range of inbred and outbred PSC lines can be stably maintained in the primed pluripotent state (epiblast stem cells -- EpiSCs) and establish the contribution of genetic variation to phenotypic differences in gene regulation and directed differentiation. Using pooled in vitro fertilization, we generate and characterize a genetic reference panel of Diversity Outbred PSCs (n = 230). Finally, we demonstrate the feasibility of pooled culture of Diversity Outbred EpiSCs as "cell villages", which can facilitate the differentiation of large numbers of EpiSC lines for forward genetic screens. These data can complement and inform similar efforts within the stem cell biology and human genetics communities to model the impact of natural genetic variation on phenotypic variation and disease-risk.
Collapse
Affiliation(s)
- Rachel A. Glenn
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cell and Developmental Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Stephanie C. Do
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Emily K. Corrigan
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Present address: Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA and Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Laura Santini
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel Medina-Cano
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarah Singer
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hyein Cho
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jing Liu
- Mouse Genetics Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karl Broman
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI USA
| | | | | | - Richard Koche
- Center for Epigenetics Research, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yasuhide Furuta
- Mouse Genetics Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meik Kunz
- The Bioinformatics CRO, Sanford Florida, 32771 USA
| | - Thomas Vierbuchen
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
46
|
Holt EA, Tyler A, Lakusta-Wong T, Lahue KG, Hankes KC, Teuscher C, Lynch RM, Ferris MT, Mahoney JM, Krementsov DN. Probing the basis of disease heterogeneity in multiple sclerosis using genetically diverse mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597205. [PMID: 38895248 PMCID: PMC11185616 DOI: 10.1101/2024.06.03.597205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Multiple sclerosis (MS) is a complex disease with significant heterogeneity in disease course and progression. Genetic studies have identified numerous loci associated with MS risk, but the genetic basis of disease progression remains elusive. To address this, we leveraged the Collaborative Cross (CC), a genetically diverse mouse strain panel, and experimental autoimmune encephalomyelitis (EAE). The thirty-two CC strains studied captured a wide spectrum of EAE severity, trajectory, and presentation, including severe-progressive, monophasic, relapsing remitting, and axial rotary (AR)-EAE, accompanied by distinct immunopathology. Sex differences in EAE severity were observed in six strains. Quantitative trait locus analysis revealed distinct genetic linkage patterns for different EAE phenotypes, including EAE severity and incidence of AR-EAE. Machine learning-based approaches prioritized candidate genes for loci underlying EAE severity ( Abcc4 and Gpc6 ) and AR-EAE ( Yap1 and Dync2h1 ). This work expands the EAE phenotypic repertoire and identifies novel loci controlling unique EAE phenotypes, supporting the hypothesis that heterogeneity in MS disease course is driven by genetic variation. Summary The genetic basis of disease heterogeneity in multiple sclerosis (MS) remains elusive. We leveraged the Collaborative Cross to expand the phenotypic repertoire of the experimental autoimmune encephalomyelitis (EAE) model of MS and identify loci controlling EAE severity, trajectory, and presentation.
Collapse
|
47
|
Gatti DM, Tyler AL, Mahoney JM, Churchill GA, Yener B, Koyuncu D, Gurcan MN, Niazi MKK, Tavolara T, Gower A, Dayao D, McGlone E, Ginese ML, Specht A, Alsharaydeh A, Tessier PA, Kurtz SL, Elkins KL, Kramnik I, Beamer G. Systems genetics uncover new loci containing functional gene candidates in Mycobacterium tuberculosis-infected Diversity Outbred mice. PLoS Pathog 2024; 20:e1011915. [PMID: 38861581 PMCID: PMC11195971 DOI: 10.1371/journal.ppat.1011915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/24/2024] [Accepted: 04/17/2024] [Indexed: 06/13/2024] Open
Abstract
Mycobacterium tuberculosis infects two billion people across the globe, and results in 8-9 million new tuberculosis (TB) cases and 1-1.5 million deaths each year. Most patients have no known genetic basis that predisposes them to disease. Here, we investigate the complex genetic basis of pulmonary TB by modelling human genetic diversity with the Diversity Outbred mouse population. When infected with M. tuberculosis, one-third develop early onset, rapidly progressive, necrotizing granulomas and succumb within 60 days. The remaining develop non-necrotizing granulomas and survive longer than 60 days. Genetic mapping using immune and inflammatory mediators; and clinical, microbiological, and granuloma correlates of disease identified five new loci on mouse chromosomes 1, 2, 4, 16; and three known loci on chromosomes 3 and 17. Further, multiple positively correlated traits shared loci on chromosomes 1, 16, and 17 and had similar patterns of allele effects, suggesting these loci contain critical genetic regulators of inflammatory responses to M. tuberculosis. To narrow the list of candidate genes, we used a machine learning strategy that integrated gene expression signatures from lungs of M. tuberculosis-infected Diversity Outbred mice with gene interaction networks to generate scores representing functional relationships. The scores were used to rank candidates for each mapped trait, resulting in 11 candidate genes: Ncf2, Fam20b, S100a8, S100a9, Itgb5, Fstl1, Zbtb20, Ddr1, Ier3, Vegfa, and Zfp318. Although all candidates have roles in infection, inflammation, cell migration, extracellular matrix remodeling, or intracellular signaling, and all contain single nucleotide polymorphisms (SNPs), SNPs in only four genes (S100a8, Itgb5, Fstl1, Zfp318) are predicted to have deleterious effects on protein functions. We performed methodological and candidate validations to (i) assess biological relevance of predicted allele effects by showing that Diversity Outbred mice carrying PWK/PhJ alleles at the H-2 locus on chromosome 17 QTL have shorter survival; (ii) confirm accuracy of predicted allele effects by quantifying S100A8 protein in inbred founder strains; and (iii) infection of C57BL/6 mice deficient for the S100a8 gene. Overall, this body of work demonstrates that systems genetics using Diversity Outbred mice can identify new (and known) QTLs and functionally relevant gene candidates that may be major regulators of complex host-pathogens interactions contributing to granuloma necrosis and acute inflammation in pulmonary TB.
Collapse
Affiliation(s)
- Daniel M. Gatti
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Anna L. Tyler
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | | | - Bulent Yener
- Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Deniz Koyuncu
- Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Metin N. Gurcan
- Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - MK Khalid Niazi
- Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - Thomas Tavolara
- Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - Adam Gower
- Clinical and Translational Science Institute, Boston University, Boston, Massachusetts, United States of America
| | - Denise Dayao
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Emily McGlone
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Melanie L. Ginese
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Aubrey Specht
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Anas Alsharaydeh
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Philipe A. Tessier
- Department of Microbiology and Immunology, Laval University School of Medicine, Quebec, Canada
| | - Sherry L. Kurtz
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Karen L. Elkins
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Igor Kramnik
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, United States of America
| | - Gillian Beamer
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| |
Collapse
|
48
|
Dillard LJ, Calabrese GM, Mesner LD, Farber CR. Cell type-specific network analysis in Diversity Outbred mice identifies genes potentially responsible for human bone mineral density GWAS associations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.594981. [PMID: 38826475 PMCID: PMC11142079 DOI: 10.1101/2024.05.20.594981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Genome-wide association studies (GWASs) have identified many sources of genetic variation associated with bone mineral density (BMD), a clinical predictor of fracture risk and osteoporosis. Aside from the identification of causal genes, other difficult challenges to informing GWAS include characterizing the roles of predicted causal genes in disease and providing additional functional context, such as the cell type predictions or biological pathways in which causal genes operate. Leveraging single-cell transcriptomics (scRNA-seq) can assist in informing BMD GWAS by linking disease-associated variants to genes and providing a cell type context for which these causal genes drive disease. Here, we use large-scale scRNA-seq data from bone marrow-derived stromal cells cultured under osteogenic conditions (BMSC-OBs) from Diversity Outbred (DO) mice to generate cell type-specific networks and contextualize BMD GWAS-implicated genes. Using trajectories inferred from the scRNA-seq data, we identify networks enriched with genes that exhibit the most dynamic changes in expression across trajectories. We discover 21 network driver genes, which are likely to be causal for human BMD GWAS associations that colocalize with expression/splicing quantitative trait loci (eQTL/sQTL). These driver genes, including Fgfrl1 and Tpx2, along with their associated networks, are predicted to be novel regulators of BMD via their roles in the differentiation of mesenchymal lineage cells. In this work, we showcase the use of single-cell transcriptomics from mouse bone-relevant cells to inform human BMD GWAS and prioritize genetic targets with potential causal roles in the development of osteoporosis.
Collapse
Affiliation(s)
- Luke J Dillard
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA 22908
| | - Gina M Calabrese
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA 22908
| | - Larry D Mesner
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA 22908
- Department of Public Health Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22908
| | - Charles R Farber
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA 22908
- Department of Public Health Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22908
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
49
|
Nagarajan A, Scoggin K, Gupta J, Aminian M, Adams LG, Kirby M, Threadgill D, Andrews-Polymenis H. Collaborative Cross mice have diverse phenotypic responses to infection with Methicillin-resistant Staphylococcus aureus USA300. PLoS Genet 2024; 20:e1011229. [PMID: 38696518 PMCID: PMC11108197 DOI: 10.1371/journal.pgen.1011229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/21/2024] [Accepted: 03/18/2024] [Indexed: 05/04/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is an opportunistic pathogen causing diseases ranging from mild skin infections to life threatening conditions, including endocarditis, pneumonia, and sepsis. To identify host genes modulating this host-pathogen interaction, we infected 25 Collaborative Cross (CC) mouse strains with methicillin-resistant S. aureus (MRSA) and monitored disease progression for seven days using a surgically implanted telemetry system. CC strains varied widely in their response to intravenous MRSA infection. We identified eight 'susceptible' CC strains with high bacterial load, tissue damage, and reduced survival. Among the surviving strains, six with minimal colonization were classified as 'resistant', while the remaining six tolerated higher organ colonization ('tolerant'). The kidney was the most heavily colonized organ, but liver, spleen and lung colonization were better correlated with reduced survival. Resistant strains had higher pre-infection circulating neutrophils and lower post-infection tissue damage compared to susceptible and tolerant strains. We identified four CC strains with sexual dimorphism: all females survived the study period while all males met our euthanasia criteria earlier. In these CC strains, males had more baseline circulating monocytes and red blood cells. We identified several CC strains that may be useful as new models for endocarditis, myocarditis, pneumonia, and resistance to MRSA infection. Quantitative Trait Locus (QTL) analysis identified two significant loci, on Chromosomes 18 and 3, involved in early susceptibility and late survival after infection. We prioritized Npc1 and Ifi44l genes as the strongest candidates influencing survival using variant analysis and mRNA expression data from kidneys within these intervals.
Collapse
Affiliation(s)
- Aravindh Nagarajan
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - Kristin Scoggin
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Jyotsana Gupta
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - Manuchehr Aminian
- Department of Mathematics, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Mathematics and Statistics, California State Polytechnic University, Pomona, California, United States of America
| | - L. Garry Adams
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Michael Kirby
- Department of Mathematics, Colorado State University, Fort Collins, Colorado, United States of America
| | - David Threadgill
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, United States of America
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas, United States of America
- Department of Biochemistry & Biophysics and Department of Nutrition, Texas A&M University, College Station, Texas, United States of America
| | - Helene Andrews-Polymenis
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
50
|
Henry AR, Miller ND, Spalding EP. QTL for the Kinematic Traits That Define the Arabidopsis Root Elongation Zone and Their Relationship to Gravitropism. PLANTS (BASEL, SWITZERLAND) 2024; 13:1189. [PMID: 38732404 PMCID: PMC11085590 DOI: 10.3390/plants13091189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024]
Abstract
Cell expansion in a discrete region called the elongation zone drives root elongation. Analyzing time lapse images can quantify the expansion in kinematic terms as if it were fluid flow. We used horizontal microscopes to collect images from which custom software extracted the length of the elongation zone, the peak relative elemental growth rate (REGR) within it, the axial position of the REGR peak, and the root elongation rate. Automation enabled these kinematic traits to be measured in 1575 Arabidopsis seedlings representing 162 recombinant inbred lines (RILs) derived from a cross of Cvi and Ler ecotypes. We mapped ten quantitative trait loci (QTL), affecting the four kinematic traits. Three QTL affected two or more traits in these vertically oriented seedlings. We compared this genetic architecture with that previously determined for gravitropism using the same RIL population. The major QTL peaks for the kinematic traits did not overlap with the gravitropism QTL. Furthermore, no single kinematic trait correlated with quantitative descriptors of the gravitropism response curve across this population. In addition to mapping QTL for growth zone traits, this study showed that the size and shape of the elongation zone may vary widely without affecting the differential growth induced by gravity.
Collapse
Affiliation(s)
| | | | - Edgar P. Spalding
- Department of Botany, University of Wisconsin, Madison, WI 53706, USA; (A.R.H.); (N.D.M.)
| |
Collapse
|