Logo: to the web site of Uppsala University

uu.sePublications from Uppsala University
Change search
Link to record
Permanent link

Direct link
Publications (10 of 178) Show all publications
Sehlin, D., Roshanbin, S., Zachrisson, O., Ingelsson, M. & Syvänen, S. (2025). A brain-penetrant bispecific antibody lowers oligomeric alpha-synuclein and activates microglia in a mouse model of alpha-synuclein pathology. Neurotherapeutics, 22(2), Article ID e00510.
Open this publication in new window or tab >>A brain-penetrant bispecific antibody lowers oligomeric alpha-synuclein and activates microglia in a mouse model of alpha-synuclein pathology
Show others...
2025 (English)In: Neurotherapeutics, ISSN 1933-7213, E-ISSN 1878-7479, Vol. 22, no 2, article id e00510Article in journal (Refereed) Published
Abstract [en]

Parkinson's disease (PD) is characterized by a progressive loss of dopaminergic neurons, linked to aggregation of alpha-synuclein (αSYN) into Lewy bodies. Current treatments are symptomatic and do not halt or reverse the neurodegeneration. Immunotherapy targeting aggregated αSYN shows potential, but therapeutic efficacy is limited by poor brain penetration of antibodies. We developed a bispecific antibody, RmAb38E2-scFv8D3, based on αSYN oligomer selective RmAb38E2 fused to a transferrin receptor (TfR)-binding domain to enhance brain delivery. Both RmAb38E2 and RmAb38E2-scFv8D3 showed higher affinity for αSYN oligomers than for monomers or fibrils. In vivo, RmAb38E2-scFv8D3 exhibited higher brain and lower blood concentrations compared to RmAb38E2, suggesting a better brain uptake and reduced peripheral exposure for the bispecific antibody. Treatment over five days of 3–4 months old transgenic L61 mice, which overexpress human αSYN, with three doses of RmAb38E2-scFv8D3 reduced brain αSYN oligomer levels and increased microglial activation, as indicated by elevated soluble TREM2 levels. Treatment with the monospecific RmAb38E2, however, showed no significant effect compared to PBS. This study demonstrates that TfR-mediated delivery enhances the therapeutic potential of αSYN-targeted immunotherapy by resulting in a higher concentration and a more uniform distribution of antibodies in the brain. The use of bispecific antibodies offers a promising strategy to improve the efficacy of antibody therapies in PD and other α-synucleinopathies.

Place, publisher, year, edition, pages
Elsevier, 2025
National Category
Neurosciences
Identifiers
urn:nbn:se:uu:diva-547032 (URN)10.1016/j.neurot.2024.e00510 (DOI)001438378500001 ()39676023 (PubMedID)2-s2.0-85212240015 (Scopus ID)
Available from: 2025-01-14 Created: 2025-01-14 Last updated: 2025-05-14Bibliographically approved
Jakalski, M., Bruhn-Olszewska, B., Rychlicka-Buniowska, E., Davies, H., Sarkisyan, D., Siedlar, M., . . . Dumanski, J. P. (2025). DNA methylation patterns contribute to changes of cellular differentiation pathways in leukocytes with LOY from patients with Alzheimers disease. Cellular and Molecular Life Sciences (CMLS), 82(1), Article ID 93.
Open this publication in new window or tab >>DNA methylation patterns contribute to changes of cellular differentiation pathways in leukocytes with LOY from patients with Alzheimers disease
Show others...
2025 (English)In: Cellular and Molecular Life Sciences (CMLS), ISSN 1420-682X, E-ISSN 1420-9071, Vol. 82, no 1, article id 93Article in journal (Refereed) Published
Abstract [en]

Alzheimer's disease (AD) is a common and increasing societal problem due to the extending human lifespan. In males, loss of chromosome Y (LOY) in leukocytes is strongly associated with AD. We studied here DNA methylation and RNA expression in sorted monocytes and granulocytes with and without LOY from male AD patients. Through multi-omic analysis, we identified new candidate genes along with those previously associated with AD. Global analyses of DNA methylation in samples with LOY vs. normal state showed that hypomethylation dominated both in granulocytes and monocytes. Our findings highlight LOY-related differences in DNA methylation that occur in gene regulatory regions. Specifically, we observed alterations in key genes involved in leukocyte differentiation: FLI1, involved in early hematopoiesis; RUNX1, essential for blood cell development; RARA, regulating gene expression in response to retinoic acid; CANX, crucial for protein folding; CEBPB, a transcription factor important for immune responses; and MYADM, implicated in cell adhesion and migration. Moreover, protein-protein interaction analysis in granulocytes identified that products of two of these genes, CANX and CEBPB, are key hub proteins. This research underscores the potential of multi-omic approach in pure hematopoietic cell populations to uncover the molecular underpinnings of AD. Finally, our results link previous analysis showing impact of LOY on leukocyte differentiation, LOY-associated transcriptional dysregulation and GWAS studies of LOY.

Place, publisher, year, edition, pages
Springer, 2025
Keywords
DNA methylation, CpG dinucleotide methylation, Loss of chromosome Y, Alzheimer's disease, Gene expression regulation
National Category
Medical Genetics and Genomics
Identifiers
urn:nbn:se:uu:diva-552038 (URN)10.1007/s00018-025-05618-8 (DOI)001432012200001 ()39998604 (PubMedID)2-s2.0-85218878235 (Scopus ID)
Note

Marcin Jąkalski and Bożena Bruhn-Olszewska shared first authors.

Available from: 2025-03-11 Created: 2025-03-11 Last updated: 2025-03-11Bibliographically approved
Klingstedt, T., Shirani, H., Parvin, F., Nyström, S., Hammarström, P., Graff, C., . . . Nilsson, K. P. (2025). Dual-ligand fluorescence microscopy enables chronological and spatial histological assignment of distinct amyloid-β deposits. Journal of Biological Chemistry, 301(1), Article ID 108032.
Open this publication in new window or tab >>Dual-ligand fluorescence microscopy enables chronological and spatial histological assignment of distinct amyloid-β deposits
Show others...
2025 (English)In: Journal of Biological Chemistry, ISSN 0021-9258, E-ISSN 1083-351X, Vol. 301, no 1, article id 108032Article in journal (Refereed) Published
Abstract [en]

Different types of deposits comprised of amyloid-β (Aβ) peptides are one of the pathological hallmarks of Alzheimer’s disease (AD) and novel methods that enable identification of a diversity of Aβ deposits during the AD continuum are essential for understanding the role of these aggregates during the pathogenesis. Herein, different combinations of five fluorescent thiophene-based ligands were used for detection of Aβ deposits in brain tissue sections from transgenic mouse models with aggregated Aβ pathology, as well as brain tissue sections from patients affected by sporadic or dominantly inherited AD. When analyzing the sections with fluorescence microscopy, distinct ligand staining patterns related to the transgenic mouse model or to the age of the mice were observed. Likewise, specific staining patterns of different Aβ deposits were revealed for sporadic versus dominantly inherited AD, as well as for distinct brain regions in sporadic AD. Thus, by using dual-staining protocols with multiple combinations of fluorescent ligands, a chronological and spatial histological designation of different Aβ deposits could be achieved. This study demonstrates the potential of our approach for resolving the role and presence of distinct Aβ aggregates during the AD continuum and pinpoints the necessity of using multiple ligands to obtain an accurate assignment of different Aβ deposits in the neuropathological evaluation of AD, as well as when evaluating therapeutic strategies targeting Aβ aggregates.

Place, publisher, year, edition, pages
Elsevier, 2025
Keywords
Alzheimer’s disease, amyloid-β, protein aggregation, fluorescence, microscopy
National Category
Clinical Laboratory Medicine Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-547600 (URN)10.1016/j.jbc.2024.108032 (DOI)001392861000001 ()39615691 (PubMedID)2-s2.0-85212576574 (Scopus ID)
Funder
Swedish Research Council, 2016-00748Swedish Research Council, 2021-01083Swedish Research Council, 2021-03524Swedish Research Council, 2023-03275Swedish Research Council, 2023-03931NIH (National Institutes of Health), 2RF1NS110437-06The Swedish Brain Foundation, FO2022-0072The Swedish Brain Foundation, FO2020-0207The Swedish Brain Foundation, ALZ2019-0004The Swedish Brain Foundation, ALZ2022-0004Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse
Available from: 2025-01-17 Created: 2025-01-17 Last updated: 2025-01-17Bibliographically approved
Tartaglia, M. C. & Ingelsson, M. (2025). Molecular Therapeutics in Development to Treat Alzheimer's Disease. Molecular Diagnosis & Therapy, 29(1), 9-24
Open this publication in new window or tab >>Molecular Therapeutics in Development to Treat Alzheimer's Disease
2025 (English)In: Molecular Diagnosis & Therapy, ISSN 1177-1062, E-ISSN 1179-2000, Vol. 29, no 1, p. 9-24Article in journal (Refereed) Published
Abstract [en]

Until recently, only symptomatic therapies, in the form of acetylcholine esterase inhibitors and NMDA-receptor antagonists, have been available for the treatment of Alzheimer’s disease. However, advancements in our understanding of the amyloid cascade hypothesis have led to a development of disease-modifying therapeutic strategies. These include immunotherapies based on an infusion of monoclonal antibodies against amyloid-β, three of which have been approved for the treatment of Alzheimer’s disease in the USA (one of them, lecanemab, has also been approved in several other countries). They all lead to a dramatic reduction of amyloid plaques in the brain, whereas their clinical effects have been more limited. Moreover, they can all lead to side effects in the form of amyloid-related imaging abnormalities. Ongoing developments aim at facilitating their administration, further improving their effects and reducing the risk for amyloid-related imaging abnormalities. Moreover, a number of anti-tau immunotherapies are in clinical trials, but none has so far shown any robust effects on symptoms or pathology. Another line of development is represented by gene therapy. To date, only antisense oligonucleotides against amyloid precursor protein/amyloid-β and tau have reached the clinical trial stage but a variety of gene editing strategies, such as clustered regularly interspaced short palindromic repeats/Cas9-mediated non-homologous end joining, base editing, and prime editing, have all shown promise on preclinical disease models. In addition, a number of other pharmacological compounds targeting a multitude of biochemical processes, believed to be centrally involved in Alzheimer’s disease, are currently being evaluated in clinical trials. This article delves into current and future perspectives on the treatment of Alzheimer’s disease, with an emphasis on immunotherapeutic and gene therapeutic strategies.

Place, publisher, year, edition, pages
Springer, 2025
National Category
Neurosciences Neurology
Identifiers
urn:nbn:se:uu:diva-555147 (URN)10.1007/s40291-024-00738-6 (DOI)001319443000001 ()39316339 (PubMedID)2-s2.0-85204794151 (Scopus ID)
Funder
Uppsala University
Note

Correction in: Molecular Diagnosis & Therapy, vol. 29, issue 1, page 143, DOI: 10.1007/s40291-024-00752-8

Available from: 2025-04-25 Created: 2025-04-25 Last updated: 2025-04-25Bibliographically approved
Hallbeck, M., Ekmark-Lewén, S., Kahle, P. J., Ingelsson, M. & Reyes, J. F. (2024). Accumulation of alpha-synuclein pathology in the liver exhibits post-translational modifications associated with Parkinson's disease. iScience, 27(12), Article ID 111448.
Open this publication in new window or tab >>Accumulation of alpha-synuclein pathology in the liver exhibits post-translational modifications associated with Parkinson's disease
Show others...
2024 (English)In: iScience, E-ISSN 2589-0042, Vol. 27, no 12, article id 111448Article in journal (Refereed) Published
Abstract [en]

Accumulating evidence demonstrates that alpha-synuclein (α-syn) pathology associated with Parkinson’s disease (PD) is not limited to the brain, as it also appears in a select number of peripheral tissues including the liver. In this study, we identified a number of PD-associated α-syn post-translational modifications in the livers of (Thy-1)-h[A30P] mice, a mouse model of familial PD expressing human α-syn harboring the A30P mutation driven by a neuron-specific promoter. Ex vivo, we also demonstrate that human hepatocytes induce post-translational modifications following α-syn fibrillar (PFF) treatment. Moreover, such cells also degrade PFFs over time, whereas oligomeric assemblies are more resistant to degradation, but this process can be enhanced by autophagy stimulators. Collectively, our findings suggest that pathological α-syn is transported to the liver in a modified state or is modified upon arrival, which facilitates its clearance and detoxification, pointing to a role for the liver in the degradation of PD-associated pathology.

Place, publisher, year, edition, pages
Elsevier, 2024
National Category
Neurosciences
Identifiers
urn:nbn:se:uu:diva-546196 (URN)10.1016/j.isci.2024.111448 (DOI)001375739200001 ()39720536 (PubMedID)2-s2.0-85211149374 (Scopus ID)
Funder
Swedish Research Council, 2019-01016The Swedish Brain FoundationThe Dementia Association - The National Association for the Rights of the DementedParkinsonfondenÅhlén-stiftelsenÅke Wiberg Foundation
Available from: 2025-01-09 Created: 2025-01-09 Last updated: 2025-01-09Bibliographically approved
Pagnon de la Vega, M., Syvänen, S., Giedraitis, V., Hooley, M., Konstantinidis, E., Meier, S. R., . . . Sehlin, D. (2024). Altered amyloid-β structure markedly reduces gliosis in the brain of mice harboring the Uppsala APP deletion. Acta neuropathologica communications, 12(1), Article ID 22.
Open this publication in new window or tab >>Altered amyloid-β structure markedly reduces gliosis in the brain of mice harboring the Uppsala APP deletion
Show others...
2024 (English)In: Acta neuropathologica communications, E-ISSN 2051-5960, Vol. 12, no 1, article id 22Article in journal (Refereed) Published
Abstract [en]

Deposition of amyloid beta (Aβ) into plaques is a major hallmark of Alzheimer’s disease (AD). Different amyloid precursor protein (APP) mutations cause early-onset AD by altering the production or aggregation properties of Aβ. We recently identified the Uppsala APP mutation (APPUpp), which causes Aβ pathology by a triple mechanism: increased β-secretase and altered α-secretase APP cleavage, leading to increased formation of a unique Aβ conformer that rapidly aggregates and deposits in the brain. The aim of this study was to further explore the effects of APPUpp in a transgenic mouse model (tg-UppSwe), expressing human APP with the APPUpp mutation together with the APPSwe mutation. Aβ pathology was studied in tg-UppSwe brains at different ages, using ELISA and immunohistochemistry. In vivo PET imaging with three different PET radioligands was conducted in aged tg-UppSwe mice and two other mouse models; tg-ArcSwe and tg-Swe. Finally, glial responses to Aβ pathology were studied in cell culture models and mouse brain tissue, using ELISA and immunohistochemistry. Tg-UppSwe mice displayed increased β-secretase cleavage and suppressed α-secretase cleavage, resulting in AβUpp42 dominated diffuse plaque pathology appearing from the age of 5–6 months. The γ-secretase cleavage was not affected. Contrary to tg-ArcSwe and tg-Swe mice, tg-UppSwe mice were [11C]PiB-PET negative. Antibody-based PET with the 3D6 ligand visualized Aβ pathology in all models, whereas the Aβ protofibril selective mAb158 ligand did not give any signals in tg-UppSwe mice. Moreover, unlike the other two models, tg-UppSwe mice displayed a very faint glial response to the Aβ pathology. The tg-UppSwe mouse model thus recapitulates several pathological features of the Uppsala APP mutation carriers. The presumed unique structural features of AβUpp42 aggregates were found to affect their interaction with anti-Aβ antibodies and profoundly modify the Aβ-mediated glial response, which may be important aspects to consider for further development of AD therapies.

Place, publisher, year, edition, pages
BioMed Central (BMC), 2024
Keywords
Alzheimer's disease (AD), Amyloid precursor protein (APP), Amyloid-beta (A beta), PET imaging, Microglia, Astrocytes, Immunotherapy
National Category
Neurosciences Neurology
Identifiers
urn:nbn:se:uu:diva-523728 (URN)10.1186/s40478-024-01734-x (DOI)001158145500001 ()38317196 (PubMedID)
Funder
Knut and Alice Wallenberg FoundationUppsala UniversitySwedish Research Council, 2016‑02120Swedish Research Council, 2021‑01083Swedish Research Council, 2021‑03524AlzheimerfondenThe Swedish Brain FoundationTorsten Söderbergs stiftelseÅhlén-stiftelsenMagnus Bergvall FoundationStiftelsen Gamla TjänarinnorGun och Bertil Stohnes StiftelseKonung Gustaf V:s och Drottning Victorias FrimurarestiftelseStiftelsen Sigurd och Elsa Goljes minne
Note

De två sista författarna delar sistaförfattarskapet

Available from: 2024-02-26 Created: 2024-02-26 Last updated: 2024-02-26Bibliographically approved
Le Borgne, J., Amouyel, P., Andreassen, O., Frikke-Schmidt, R., Hiltunen, M., Ingelsson, M., . . . Bellenguez, C. (2024). Association of MGMT and BIN1 genes with Alzheimer's disease risk across sex and APOE ε4 status [Letter to the editor]. Alzheimer's & Dementia: Journal of the Alzheimer's Association, 20(3), 2282-2284
Open this publication in new window or tab >>Association of MGMT and BIN1 genes with Alzheimer's disease risk across sex and APOE ε4 status
Show others...
2024 (English)In: Alzheimer's & Dementia: Journal of the Alzheimer's Association, ISSN 1552-5260, E-ISSN 1552-5279, Vol. 20, no 3, p. 2282-2284Article in journal, Letter (Other academic) Published
Place, publisher, year, edition, pages
John Wiley & Sons, 2024
National Category
Neurosciences Psychiatry Public Health, Global Health and Social Medicine
Identifiers
urn:nbn:se:uu:diva-534534 (URN)10.1002/alz.13550 (DOI)001112883500001 ()38041824 (PubMedID)
Funder
The Research Council of Norway, 223273The Research Council of Norway, 283799The Research Council of Norway, 324252The Research Council of Norway, 344121
Available from: 2024-07-04 Created: 2024-07-04 Last updated: 2025-02-20Bibliographically approved
Eltom, K., Mothes, T., Libard, S., Ingelsson, M. & Erlandsson, A. (2024). Astrocytic accumulation of tau fibrils isolated from Alzheimer’s disease brains induces inflammation, cell-to-cell propagation and neuronal impairment. Acta neuropathologica communications, 12(1), Article ID 34.
Open this publication in new window or tab >>Astrocytic accumulation of tau fibrils isolated from Alzheimer’s disease brains induces inflammation, cell-to-cell propagation and neuronal impairment
Show others...
2024 (English)In: Acta neuropathologica communications, E-ISSN 2051-5960, Vol. 12, no 1, article id 34Article in journal (Refereed) Published
Abstract [en]

Accumulating evidence highlights the involvement of astrocytes in Alzheimer’s disease (AD) progression. We have previously demonstrated that human iPSC-derived astrocytes ingest and modify synthetic tau fibrils in a way that enhances their seeding efficiency. However, synthetic tau fibrils differ significantly from in vivo formed fibrils. To mimic the situation in the brain, we here analyzed astrocytes’ processing of human brain-derived tau fibrils and its consequences for cellular physiology. Tau fibrils were extracted from both AD and control brains, aiming to examine any potential differences in astrocyte response depending on the origin of fibrils. Our results show that human astrocytes internalize, but fail to degrade, both AD and control tau fibrils. Instead, pathogenic, seeding capable tau proteoforms are spread to surrounding cells via tunneling nanotubes and exocytosis. Notably, accumulation of AD tau fibrils induces a stronger reactive state in astrocytes, compared to control fibrils, evident by the augmented expression of vimentin and GFAP, as well as by an increased secretion of the pro-inflammatory cytokines IL-8 and MCP-1. Moreover, conditioned media from astrocytes with AD tau fibril deposits induce synapse and metabolic impairment in human iPSC-derived neurons. Taken together, our data suggest that the accumulation of brain-derived AD tau fibrils induces a more robust inflammatory and neurotoxic phenotype in human astrocytes, accentuating the nature of tau fibrils as an important contributing factor to inflammation and neurodegeneration in AD. 

Place, publisher, year, edition, pages
BioMed Central (BMC), 2024
Keywords
Alzheimer’s disease; tau; astrocytes; brain-derived fibrils; inflammation; neurons
National Category
Neurosciences
Identifiers
urn:nbn:se:uu:diva-523823 (URN)10.1186/s40478-024-01745-8 (DOI)001176894300001 ()38409026 (PubMedID)
Funder
Åhlén-stiftelsen, 233044The Swedish Brain Foundation, FO2022-0083Stiftelsen Gamla Tjänarinnor, 2021 − 01171O.E. och Edla Johanssons vetenskapliga stiftelseBertil and Ebon Norlin Foundation for Medical ResearchGun och Bertil Stohnes StiftelseSwedish Fund for Research Without Animal Experiments, F2022-0004Uppsala University
Available from: 2024-02-23 Created: 2024-02-23 Last updated: 2024-04-10Bibliographically approved
Wu, X., Shirani, H., Vidal, R., Ghetti, B., Ingelsson, M., Klingstedt, T. & Nilsson, K. P. (2024). Distinct Chemical Determinants are Essential for Achieving Ligands for Superior Optical Detection of Specific Amyloid-ß Deposits in Alzheimer's Disease. ChemistryOpen, 13(12), Article ID e202400186.
Open this publication in new window or tab >>Distinct Chemical Determinants are Essential for Achieving Ligands for Superior Optical Detection of Specific Amyloid-ß Deposits in Alzheimer's Disease
Show others...
2024 (English)In: ChemistryOpen, ISSN 2191-1363, Vol. 13, no 12, article id e202400186Article in journal (Refereed) Published
Abstract [en]

Aggregated forms of different proteins are common hallmarks for several neurodegenerative diseases, including Alzheimer's disease, and ligands that selectively detect specific protein aggregates are vital. Herein, we investigate the molecular requirements of thiophene-vinyl-benzothiazole based ligands to detect a specific type of A beta deposits found in individuals with dominantly inherited Alzheimer's disease caused by the Arctic APP E693G mutation. The staining of these A beta deposits was alternated when switching the terminal heterocyclic moiety attached to the thiophene-vinyl-benzothiazole scaffold. The most prevalent staining was observed for ligands having a terminal 3-methyl-1H-indazole moiety or a terminal 1,2-dimethoxybenzene moiety, verifying that specific molecular interactions between these ligands and the aggregates were necessary. The synthesis of additional thiophene-vinyl-benzothiazole ligands aided in pinpointing additional crucial chemical determinants, such as positioning of nitrogen atoms and methyl substituents, for achieving optimal staining of A beta aggregates. When combining the optimized thiophene-vinyl-benzothiazole based ligands with a conventional ligand, CN-PiB, distinct staining patterns were observed for sporadic Alzheimer's disease versus dominantly inherited Alzheimer's disease caused by the Arctic APP E693G mutation. Our findings provide chemical insights for developing novel ligands that allow for a more precise assignment of A beta deposits, and might also aid in creating novel agents for clinical imaging of distinct A beta aggregates in AD.

Place, publisher, year, edition, pages
John Wiley & Sons, 2024
Keywords
Ligands, Fluorescence, Alzheimer's disease, Amyloid beta, Protein aggregates
National Category
Neurosciences
Identifiers
urn:nbn:se:uu:diva-548392 (URN)10.1002/open.202400186 (DOI)001355428800001 ()39508558 (PubMedID)
Funder
Swedish Research Council, 2016-00748Swedish Research Council, 2023-03275The Swedish Brain Foundation
Available from: 2025-01-24 Created: 2025-01-24 Last updated: 2025-01-24Bibliographically approved
Suzuki, K., Giedraitis, V., Ingelsson, M., Lind, L., Ingelsson, E. & Zeggini, E. (2024). Genetic drivers of heterogeneity in type 2 diabetes pathophysiology. Nature, 627(8003), 347-357
Open this publication in new window or tab >>Genetic drivers of heterogeneity in type 2 diabetes pathophysiology
Show others...
2024 (English)In: Nature, ISSN 0028-0836, E-ISSN 1476-4687, Vol. 627, no 8003, p. 347-357Article in journal (Refereed) Published
Abstract [en]

Type 2 diabetes (T2D) is a heterogeneous disease that develops through diverse pathophysiological processes1,2 and molecular mechanisms that are often specific to cell type3,4. Here, to characterize the genetic contribution to these processes across ancestry groups, we aggregate genome-wide association study data from 2,535,601 individuals (39.7% not of European ancestry), including 428,452 cases of T2D. We identify 1,289 independent association signals at genome-wide significance (P < 5 × 10−8) that map to 611 loci, of which 145 loci are, to our knowledge, previously unreported. We define eight non-overlapping clusters of T2D signals that are characterized by distinct profiles of cardiometabolic trait associations. These clusters are differentially enriched for cell-type-specific regions of open chromatin, including pancreatic islets, adipocytes, endothelial cells and enteroendocrine cells. We build cluster-specific partitioned polygenic scores5 in a further 279,552 individuals of diverse ancestry, including 30,288 cases of T2D, and test their association with T2D-related vascular outcomes. Cluster-specific partitioned polygenic scores are associated with coronary artery disease, peripheral artery disease and end-stage diabetic nephropathy across ancestry groups, highlighting the importance of obesity-related processes in the development of vascular outcomes. Our findings show the value of integrating multi-ancestry genome-wide association study data with single-cell epigenomics to disentangle the aetiological heterogeneity that drives the development and progression of T2D. This might offer a route to optimize global access to genetically informed diabetes care.

Place, publisher, year, edition, pages
Springer Nature, 2024
National Category
Endocrinology and Diabetes Medical Genetics and Genomics
Identifiers
urn:nbn:se:uu:diva-533526 (URN)10.1038/s41586-024-07019-6 (DOI)001185035100001 ()38374256 (PubMedID)
Funder
EU, Horizon 2020, 101017802
Note

For complete list of authors see http://dx.doi.org/10.1038/s41586-024-07019-6

Available from: 2024-06-27 Created: 2024-06-27 Last updated: 2025-02-10Bibliographically approved
Projects
Alpha-synuclein pathogenesis - novel targets for therapy and diagnostics in Parkinson´s disease [2011-04519_VR]; Uppsala UniversityA European DNA bank for deciphering the missing heritability of Alzheimer’s disease [2015-06799_VR]; Uppsala UniversityDevelopment of gene therapy targeting amyloid-Œ≤ and Œ±-synuclein on cell and mouse models for Alzheimer’s disease and Parkinson’s disease [2018-03075_VR]; Uppsala UniversityFamilial forms of Alzheimer’s disease: Investigation of the novel APPAros mutation and continued development of CRISPR-based gene therapy [2021-02793_VR]; Uppsala University
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0001-5466-8370

Search in DiVA

Show all publications