To Örebro University

oru.seÖrebro University Publications
Change search
Link to record
Permanent link

Direct link
Bedoya Reina, Oscar
Alternative names
Publications (10 of 23) Show all publications
Mei, S., Alchahin, A. M., Embaie, B. T., Gavriliuc, I. M., Verhoeven, B. M., Zhao, T., . . . Baryawno, N. (2024). Single-cell analyses of metastatic bone marrow in human neuroblastoma reveals microenvironmental remodeling and metastatic signature. JCI Insight, 9(6), Article ID e173337.
Open this publication in new window or tab >>Single-cell analyses of metastatic bone marrow in human neuroblastoma reveals microenvironmental remodeling and metastatic signature
Show others...
2024 (English)In: JCI Insight, ISSN 2379-3708, Vol. 9, no 6, article id e173337Article in journal (Refereed) Published
Abstract [en]

Neuroblastoma is an aggressive pediatric cancer with a high rate of metastasis to the BM. Despite intensive treatments including high-dose chemotherapy, the overall survival rate for children with metastatic neuroblastoma remains dismal. Understanding the cellular and molecular mechanisms of the metastatic tumor microenvironment is crucial for developing new therapies and improving clinical outcomes. Here, we used single-cell RNA-Seq to characterize immune and tumor cell alterations in neuroblastoma BM metastases by comparative analysis with patients without metastases. Our results reveal remodeling of the immune cell populations and reprogramming of gene expression profiles in the metastatic niche. In particular, within the BM metastatic niche, we observed the enrichment of immune cells, including tumor-associated neutrophils, macrophages, and exhausted T cells, as well as an increased number of Tregs and a decreased number of B cells. Furthermore, we highlighted cell communication between tumor cells and immune cell populations, and we identified prognostic markers in malignant cells that are associated with worse clinical outcomes in 3 independent neuroblastoma cohorts. Our results provide insight into the cellular, compositional, and transcriptional shifts underlying neuroblastoma BM metastases that contribute to the development of new therapeutic strategies. 

Place, publisher, year, edition, pages
American Society For Clinical Investigation, 2024
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:oru:diva-118760 (URN)10.1172/jci.insight.173337 (DOI)001189379000001 ()38358826 (PubMedID)2-s2.0-85188761032 (Scopus ID)
Funder
Swedish Childhood Cancer FoundationSwedish Cancer SocietyThe Cancer Research Funds of RadiumhemmetWenner-Gren Foundations
Available from: 2025-01-22 Created: 2025-01-22 Last updated: 2025-01-23Bibliographically approved
Polychronopoulos, P. A., Bedoya Reina, O. & Johnsen, J. I. (2024). The Neuroblastoma Microenvironment, Heterogeneity and Immunotherapeutic Approaches. Cancers, 16(10), Article ID 1863.
Open this publication in new window or tab >>The Neuroblastoma Microenvironment, Heterogeneity and Immunotherapeutic Approaches
2024 (English)In: Cancers, ISSN 2072-6694, Vol. 16, no 10, article id 1863Article, review/survey (Refereed) Published
Abstract [en]

Neuroblastoma is a peripheral nervous system tumor that almost exclusively occurs in young children. Although intensified treatment modalities have led to increased patient survival, the prognosis for patients with high-risk disease is still around 50%, signifying neuroblastoma as a leading cause of cancer-related deaths in children. Neuroblastoma is an embryonal tumor and is shaped by its origin from cells within the neural crest. Hence, neuroblastoma usually presents with a low mutational burden and is, in the majority of cases, driven by epigenetically deregulated transcription networks. The recent development of Omic techniques has given us detailed knowledge of neuroblastoma evolution, heterogeneity, and plasticity, as well as intra- and intercellular molecular communication networks within the neuroblastoma microenvironment. Here, we discuss the potential of these recent discoveries with emphasis on new treatment modalities, including immunotherapies which hold promise for better future treatment regimens.

Place, publisher, year, edition, pages
MDPI, 2024
Keywords
Immunotherapy, neural crest, neuroblastoma, tumor microenvironment
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:oru:diva-113869 (URN)10.3390/cancers16101863 (DOI)001232657600001 ()38791942 (PubMedID)2-s2.0-85194361798 (Scopus ID)
Funder
Swedish Cancer Society, 21 1556Swedish Childhood Cancer Foundation, PR2020-0140King Gustaf V Jubilee Fund, 154048
Available from: 2024-05-27 Created: 2024-05-27 Last updated: 2024-06-11Bibliographically approved
Yuan, Y., Alzrigat, M., Rodriguez-Garcia, A., Wang, X., Bexelius, T. S., Johnsen, J. I., . . . Bedoya-Reina, O. C. (2023). Target Genes of c-MYC and MYCN with Prognostic Power in Neuroblastoma Exhibit Different Expressions during Sympathoadrenal Development. Cancers, 15(18), Article ID 4599.
Open this publication in new window or tab >>Target Genes of c-MYC and MYCN with Prognostic Power in Neuroblastoma Exhibit Different Expressions during Sympathoadrenal Development
Show others...
2023 (English)In: Cancers, ISSN 2072-6694, Vol. 15, no 18, article id 4599Article in journal (Refereed) Published
Abstract [en]

Deregulation of the MYC family of transcription factors c-MYC (encoded by MYC), MYCN, and MYCL is prevalent in most human cancers, with an impact on tumor initiation and progression, as well as response to therapy. In neuroblastoma (NB), amplification of the MYCN oncogene and over-expression of MYC characterize approximately 40% and 10% of all high-risk NB cases, respectively. However, the mechanism and stage of neural crest development in which MYCN and c-MYC contribute to the onset and/or progression of NB are not yet fully understood. Here, we hypothesized that subtle differences in the expression of MYCN and/or c-MYC targets could more accurately stratify NB patients in different risk groups rather than using the expression of either MYC gene alone. We employed an integrative approach using the transcriptome of 498 NB patients from the SEQC cohort and previously defined c-MYC and MYCN target genes to model a multigene transcriptional risk score. Our findings demonstrate that defined sets of c-MYC and MYCN targets with significant prognostic value, effectively stratify NB patients into different groups with varying overall survival probabilities. In particular, patients exhibiting a high-risk signature score present unfavorable clinical parameters, including increased clinical risk, higher INSS stage, MYCN amplification, and disease progression. Notably, target genes with prognostic value differ between c-MYC and MYCN, exhibiting distinct expression patterns in the developing sympathoadrenal system. Genes associated with poor outcomes are mainly found in sympathoblasts rather than in chromaffin cells during the sympathoadrenal development.

Place, publisher, year, edition, pages
MDPI, 2023
Keywords
Neuroblastoma, c-MYC, MYCN, prognosis, gene signature, sympathoadrenal development
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:oru:diva-118761 (URN)10.3390/cancers15184599 (DOI)001073751900001 ()37760568 (PubMedID)2-s2.0-85172761076 (Scopus ID)
Funder
Swedish Research Council, 2018-05973Swedish Childhood Cancer Foundation, PR2021-0129
Available from: 2025-01-22 Created: 2025-01-22 Last updated: 2025-01-23Bibliographically approved
Sainero-Alcolado, L., Mushtaq, M., Liaño-Pons, J., Rodriguez-Garcia, A., Yuan, Y., Liu, T., . . . Arsenian-Henriksson, M. (2022). Expression and activation of nuclear hormone receptors result in neuronal differentiation and favorable prognosis in neuroblastoma. Journal of Experimental & Clinical Cancer Research, 41(1), Article ID 226.
Open this publication in new window or tab >>Expression and activation of nuclear hormone receptors result in neuronal differentiation and favorable prognosis in neuroblastoma
Show others...
2022 (English)In: Journal of Experimental & Clinical Cancer Research, E-ISSN 1756-9966, Vol. 41, no 1, article id 226Article in journal (Refereed) Published
Abstract [en]

Background: Neuroblastoma (NB), a childhood tumor derived from the sympathetic nervous system, presents with heterogeneous clinical behavior. While some tumors regress spontaneously without medical intervention, others are resistant to therapy, associated with an aggressive phenotype. MYCN-amplification, frequently occurring in high-risk NB, is correlated with an undifferentiated phenotype and poor prognosis. Differentiation induction has been proposed as a therapeutic approach for high-risk NB. We have previously shown that MYCN maintains an undifferentiated state via regulation of the miR-17 ~ 92 microRNA cluster, repressing the nuclear hormone receptors (NHRs) estrogen receptor alpha (ERα) and the glucocorticoid receptor (GR).

Methods: Cell viability was determined by WST-1. Expression of differentiation markers was analyzed by Western blot, RT-qPCR, and immunofluorescence analysis. Metabolic phenotypes were studied using Agilent Extracellular Flux Analyzer, and accumulation of lipid droplets by Nile Red staining. Expression of angiogenesis, proliferation, and neuronal differentiation markers, and tumor sections were assessed by immunohistochemistry. Gene expression from NB patient as well as adrenal gland cohorts were analyzed using GraphPad Prism software (v.8) and GSEA (v4.0.3), while pseudo-time progression on post-natal adrenal gland cells from single-nuclei transcriptome data was computed using scVelo.

Results: Here, we show that simultaneous activation of GR and ERα potentiated induction of neuronal differentiation, reduced NB cell viability in vitro, and decreased tumor burden in vivo. This was accompanied by a metabolic reprogramming manifested by changes in the glycolytic and mitochondrial functions and in lipid droplet accumulation. Activation of the retinoic acid receptor alpha (RARα) with all-trans retinoic acid (ATRA) further enhanced the differentiated phenotype as well as the metabolic switch. Single-cell nuclei transcriptome analysis of human adrenal glands indicated a sequential expression of ERα, GR, and RARα during development from progenitor to differentiated chromaffin cells. Further, in silico analysis revealed that patients with higher combined expression of GR, ERα, and RARα mRNA levels had elevated expression of neuronal differentiation markers and a favorable outcome.

Conclusion: Together, our findings suggest that combination therapy involving activation of several NHRs could be a promising pharmacological approach for differentiation treatment of NB patients. Keywords:  Estrogen receptor α; Glucocorticoid receptor; Metabolic reprogramming; Neuroblastoma; Neuronal differentiation; Nuclear hormone receptors; Retinoic acid receptor α.

Place, publisher, year, edition, pages
BioMed Central (BMC), 2022
Keywords
Neuroblastoma, Nuclear hormone receptors, Neuronal differentiation, Metabolic reprogramming, Glucocorticoid receptor, Estrogen receptor alpha, Retinoic acid receptor alpha
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:oru:diva-118764 (URN)10.1186/s13046-022-02399-x (DOI)000826927900001 ()35850708 (PubMedID)2-s2.0-85134415315 (Scopus ID)
Funder
Swedish Cancer SocietySwedish Research CouncilSwedish Childhood Cancer FoundationThe Cancer Research Funds of RadiumhemmetKarolinska Institute
Available from: 2025-01-22 Created: 2025-01-22 Last updated: 2025-01-23Bibliographically approved
Li, S., Li, W., Yuan, J., Bullova, P., Wu, J., Zhang, X., . . . Schlisio, S. (2022). Impaired oxygen-sensitive regulation of mitochondrial biogenesis within the von Hippel–Lindau syndrome. Nature Metabolism, 4(6), 739-758
Open this publication in new window or tab >>Impaired oxygen-sensitive regulation of mitochondrial biogenesis within the von Hippel–Lindau syndrome
Show others...
2022 (English)In: Nature Metabolism, E-ISSN 2522-5812, Vol. 4, no 6, p. 739-758Article in journal (Refereed) Published
Abstract [en]

Mitochondria are the main consumers of oxygen within the cell. How mitochondria sense oxygen levels remains unknown. Here we show an oxygen-sensitive regulation of TFAM, an activator of mitochondrial transcription and replication, whose alteration is linked to tumours arising in the von Hippel-Lindau syndrome. TFAM is hydroxylated by EGLN3 and subsequently bound by the von Hippel-Lindau tumour-suppressor protein, which stabilizes TFAM by preventing mitochondrial proteolysis. Cells lacking wild-type VHL or in which EGLN3 is inactivated have reduced mitochondrial mass. Tumorigenic VHL variants leading to different clinical manifestations fail to bind hydroxylated TFAM. In contrast, cells harbouring the Chuvash polycythaemia VHLR200W mutation, involved in hypoxia-sensing disorders without tumour development, are capable of binding hydroxylated TFAM. Accordingly, VHL-related tumours, such as pheochromocytoma and renal cell carcinoma cells, display low mitochondrial content, suggesting that impaired mitochondrial biogenesis is linked to VHL tumorigenesis. Finally, inhibiting proteolysis by targeting LONP1 increases mitochondrial content in VHL-deficient cells and sensitizes therapy-resistant tumours to sorafenib treatment. Our results offer pharmacological avenues to sensitize therapy-resistant VHL tumours by focusing on the mitochondria.

Place, publisher, year, edition, pages
Nature Publishing Group, 2022
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:oru:diva-118765 (URN)10.1038/s42255-022-00593-x (DOI)000817322200011 ()35760869 (PubMedID)2-s2.0-85132952541 (Scopus ID)
Funder
Wellcome trust, 208402/Z/17/Z
Note

Publisher Correction: Impaired oxygen-sensitive regulation of mitochondrial biogenesis within the von Hippel–Lindau syndrome. Li, S., Li, W., Yuan, J. et al. Nat Metab 4, 1421 (2022). https://doi.org/10.1038/s42255-022-00651-4

Available from: 2025-01-22 Created: 2025-01-22 Last updated: 2025-01-23Bibliographically approved
Alkasalias, T., Zhang, J., Madapura, H., Dalarun, B., Bedoya Reina, O., Lewensohn, R., . . . Laín, S. (2022). Proof-of-principle studies on a strategy to enhance nucleotide imbalance specifically in cancer cells. Cell Death Discovery, 8(1), Article ID 464.
Open this publication in new window or tab >>Proof-of-principle studies on a strategy to enhance nucleotide imbalance specifically in cancer cells
Show others...
2022 (English)In: Cell Death Discovery, E-ISSN 2058-7716, Vol. 8, no 1, article id 464Article in journal (Refereed) Published
Abstract [en]

Highly specific and potent inhibitors of dihydroorotate dehydrogenase (DHODH), an essential enzyme of the de novo pyrimidine ribonucleotide synthesis pathway, are in clinical trials for autoimmune diseases, viral infections and cancer. However, because DHODH inhibitors (DHODHi) are immunosuppressants they may reduce the anticancer activity of the immune system. Therefore, there may be a need to improve the therapeutic index of DHODHi in cancer patients. The aim of this study was to find strategies to protect activated T cells from DHODHi and to identify cancer types hypersensitive to these inhibitors. First, we observed that like uridine supplementation, adding cytidine to the culture medium protects T cells from DHODH blockage. Next, we identified tumor types with altered expression of pyrimidine ribonucleotide synthesis enzymes. In this regard, we detected that the expression of cytidine deaminase (CDA), which converts cytidine into uridine, is low in an important proportion of cancer cell lines and consistently low in neuroblastoma samples and in cell lines from neuroblastoma and small cell lung carcinoma. This suggested that in the presence of a DHODHi, an excess of cytidine would be deleterious for low CDA expressing cancer cell lines. We show that this was the case (as could be seen almost immediately after treatment) when cells were cultured with fetal bovine serum but, was significantly less evident when cultures contained human serum. One interesting feature of CDA is that aside from acting intracellularly, it is also present in human plasma/serum. Altogether, experiments using recombinant CDA, human serum, pharmacologic inhibition of CDA and T cell/cancer cell co-cultures suggest that the therapeutic index of DHODHi could be improved by selecting patients with low-CDA expressing cancers in combination with strategies to increase cytidine or the cytidine/uridine ratio in the extracellular environment. Collectively, this proof-of-principle study warrants the discovery of agents to deplete extracellular CDA.

Place, publisher, year, edition, pages
Springer Nature, 2022
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:oru:diva-118762 (URN)10.1038/s41420-022-01254-4 (DOI)000887921900002 ()36424385 (PubMedID)2-s2.0-85142479279 (Scopus ID)
Funder
Karolinska Institute
Note

Correction: Proof-of-principle studies on a strategy to enhance nucleotide imbalance specifically in cancer cells. Alkasalias, T., Zhang, J., Madapura, H. et al. Cell Death Discov. 8, 472 (2022). https://doi.org/10.1038/s41420-022-01275-z

Available from: 2025-01-22 Created: 2025-01-22 Last updated: 2025-01-23Bibliographically approved
Shi, Y., Yuan, J., Rraklli, V., Maxymovitz, E., Cipullo, M., Liu, M., . . . Holmberg, J. (2021). Aberrant splicing in neuroblastoma generates RNA-fusion transcripts and provides vulnerability to spliceosome inhibitors. Nucleic Acids Research, 49(5), 2509-2521
Open this publication in new window or tab >>Aberrant splicing in neuroblastoma generates RNA-fusion transcripts and provides vulnerability to spliceosome inhibitors
Show others...
2021 (English)In: Nucleic Acids Research, ISSN 0305-1048, E-ISSN 1362-4962, Vol. 49, no 5, p. 2509-2521Article in journal (Refereed) Published
Abstract [en]

The paucity of recurrent mutations has hampered efforts to understand and treat neuroblastoma. Alternative splicing and splicing-dependent RNA-fusions represent mechanisms able to increase the gene product repertoire but their role in neuroblastoma remains largely unexplored. Here we investigate the presence and possible roles of aberrant splicing and splicing-dependent RNA-fusion transcripts in neuroblastoma. In addition, we attend to establish whether the spliceosome can be targeted to treat neuroblastoma. Through analysis of RNA-sequenced neuroblastoma we show that elevated expression of splicing factors is a strong predictor of poor clinical outcome. Furthermore, we identified >900 primarily intrachromosomal fusions containing canonical splicing sites. Fusions included transcripts from well-known oncogenes, were enriched for proximal genes and in chromosomal regions commonly gained or lost in neuroblastoma. As a proof-of-principle that these fusions can generate altered gene products, we characterized a ZNF451-BAG2 fusion, producing a truncated BAG2-protein which inhibited retinoic acid induced differentiation. Spliceosome inhibition impeded neuroblastoma fusion expression, induced apoptosis and inhibited xenograft tumor growth. Our findings elucidate a splicing-dependent mechanism generating altered gene products in neuroblastoma and show that the spliceosome is a potential target for clinical intervention.

Place, publisher, year, edition, pages
Oxford University Press, 2021
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:oru:diva-118767 (URN)10.1093/nar/gkab054 (DOI)000637321700013 ()33555349 (PubMedID)2-s2.0-85103229013 (Scopus ID)
Funder
Knut and Alice Wallenberg FoundationSwedish Research CouncilThe Swedish Brain Foundation
Available from: 2025-01-22 Created: 2025-01-22 Last updated: 2025-01-23Bibliographically approved
Bedoya Reina, O. & Schlisio, S. (2021). Chromaffin Cells with Sympathoblast Signature: Too Similar to Keep Apart? [Letter to the editor]. Cancer Cell, 39(2), 134-135
Open this publication in new window or tab >>Chromaffin Cells with Sympathoblast Signature: Too Similar to Keep Apart?
2021 (English)In: Cancer Cell, ISSN 1535-6108, E-ISSN 1878-3686, Vol. 39, no 2, p. 134-135Article in journal, Letter (Refereed) Published
Place, publisher, year, edition, pages
Cell Press, 2021
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:oru:diva-118768 (URN)10.1016/j.ccell.2020.12.009 (DOI)000627120000005 ()33385330 (PubMedID)2-s2.0-85099667820 (Scopus ID)
Funder
Swedish Research Council Formas, 2016-07213Swedish Research Council Formas, 2018-05973Swedish Research Council FormasSwedish Cancer SocietyKnut and Alice Wallenberg Foundation
Available from: 2025-01-22 Created: 2025-01-22 Last updated: 2025-01-23Bibliographically approved
Bedoya Reina, O., Li, W., Arceo, M., Plescher, M., Bullova, P., Pui, H., . . . Schlisio, S. (2021). Single-nuclei transcriptomes from human adrenal gland reveal distinct cellular identities of low and high-risk neuroblastoma tumors. Nature Communications, 12(1), Article ID 5309.
Open this publication in new window or tab >>Single-nuclei transcriptomes from human adrenal gland reveal distinct cellular identities of low and high-risk neuroblastoma tumors
Show others...
2021 (English)In: Nature Communications, E-ISSN 2041-1723, Vol. 12, no 1, article id 5309Article in journal (Refereed) Published
Abstract [en]

Childhood neuroblastoma has a remarkable variability in outcome. Age at diagnosis is one of the most important prognostic factors, with children less than 1 year old having favorable outcomes. Here we study single-cell and single-nuclei transcriptomes of neuroblastoma with different clinical risk groups and stages, including healthy adrenal gland. We compare tumor cell populations with embryonic mouse sympatho-adrenal derivatives, and post-natal human adrenal gland. We provide evidence that low and high-risk neuroblastoma have different cell identities, representing two disease entities. Low-risk neuroblastoma presents a transcriptome that resembles sympatho- and chromaffin cells, whereas malignant cells enriched in high-risk neuroblastoma resembles a subtype of TRKB+ cholinergic progenitor population identified in human post-natal gland. Analyses of these populations reveal different gene expression programs for worst and better survival in correlation with age at diagnosis. Our findings reveal two cellular identities and a composition of human neuroblastoma tumors reflecting clinical heterogeneity and outcome.

Place, publisher, year, edition, pages
Springer Nature, 2021
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:oru:diva-118766 (URN)10.1038/s41467-021-24870-7 (DOI)000694655700013 ()34493726 (PubMedID)2-s2.0-85114593900 (Scopus ID)
Funder
Swedish Research Council, 2016-07213Swedish Research Council, 2018-05973Swedish Childhood Cancer FoundationKnut and Alice Wallenberg FoundationEU, European Research CouncilSwedish Foundation for Strategic ResearchEU, European Research CouncilSwedish Cancer Society
Note

Funding Agencies:

Swedish Research Council 

 Swedish Childhood Cancer Fund

 Swedish Cancer Society

 Knut & Alice Wallenberg Foundation

 ParaDiff Foundation

 European Research Council (ERC)

 Swedish Foundation for Strategic Research

 European Research Council (ERC)

 Paradifference Foundation

 Bertil Hallsten Research Foundation

 Cancer Foundation in Sweden

Available from: 2025-01-22 Created: 2025-01-22 Last updated: 2025-01-23Bibliographically approved
Kitchen, S. A., Ratan, A., Bedoya Reina, O. C., Burhans, R., Fogarty, N. D., Miller, W. & Baums, I. B. (2019). Genomic Variants Among Threatened Acropora Corals. G3: Genes, Genomes, Genetics, 9(5), 1633-1646
Open this publication in new window or tab >>Genomic Variants Among Threatened Acropora Corals
Show others...
2019 (English)In: G3: Genes, Genomes, Genetics, E-ISSN 2160-1836, Vol. 9, no 5, p. 1633-1646Article in journal (Refereed) Published
Abstract [en]

Genomic sequence data for non-model organisms are increasingly available requiring the development of efficient and reproducible workflows. Here, we develop the first genomic resources and reproducible workflows for two threatened members of the reef-building coral genus Acropora. We generated genomic sequence data from multiple samples of the Caribbean A. cervicornis (staghorn coral) and A. palmata (elkhorn coral), and predicted millions of nucleotide variants among these two species and the Pacific A. digitifera. A subset of predicted nucleotide variants were verified using restriction length polymorphism assays and proved useful in distinguishing the two Caribbean acroporids and the hybrid they form (“A. prolifera”). Nucleotide variants are freely available from the Galaxy server (usegalaxy.org), and can be analyzed there with computational tools and stored workflows that require only an internet browser. We describe these data and some of the analysis tools, concentrating on fixed differences between A. cervicornis and A. palmata. In particular, we found that fixed amino acid differences between these two species were enriched in proteins associated with development, cellular stress response, and the host’s interactions with associated microbes, for instance in the ABC transporters and superoxide dismutase. Identified candidate genes may underlie functional differences in how these threatened species respond to changing environments. Users can expand the presented analyses easily by adding genomic data from additional species, as they become available.

Place, publisher, year, edition, pages
The Genetics Society, 2019
Keywords
Coral, Caribbean, single nucleotide polymorphism, population genomics, Galaxy
National Category
Genetics and Genomics
Identifiers
urn:nbn:se:oru:diva-118770 (URN)10.1534/g3.119.400125 (DOI)000467271400032 ()30914426 (PubMedID)2-s2.0-85065798446 (Scopus ID)
Note

Funding Agency:

National Science Foundation (NSF)

Available from: 2025-01-22 Created: 2025-01-22 Last updated: 2025-01-23Bibliographically approved
Organisations

Search in DiVA

Show all publications