kth.sePublications
Change search
Link to record
Permanent link

Direct link
Publications (10 of 35) Show all publications
Shi, M., Shi, M., Karlsson, M., Alvez, M. B., Jin, H., Yuan, M., . . . et al., . (2025). A resource for whole-body gene expression map of human tissues based on integration of single cell and bulk transcriptomics. Genome Biology, 26(1), Article ID 152.
Open this publication in new window or tab >>A resource for whole-body gene expression map of human tissues based on integration of single cell and bulk transcriptomics
Show others...
2025 (English)In: Genome Biology, ISSN 1465-6906, E-ISSN 1474-760X, Vol. 26, no 1, article id 152Article in journal (Refereed) Published
Abstract [en]

New technologies enable single-cell transcriptome analysis, mapping genome-wide expression across the human body. Here, we present an extended analysis of protein-coding genes in all major human tissues and organs, combining single-cell and bulk transcriptomics. To enhance transcriptome depth, 31 tissues were analyzed using a pooling method, identifying 557 unique cell clusters, manually annotated by marker gene expression. Genes were classified by body-wide expression and validated through antibody-based profiling. All results are available in the updated open-access Single Cell Type section of the Human Protein Atlas for genome-wide exploration of genes, proteins, and their spatial distribution in cells.

Place, publisher, year, edition, pages
Springer Nature, 2025
Keywords
Cell type classification, Gene expression mapping, Human Protein Atlas, Single-cell
National Category
Bioinformatics and Computational Biology Cell and Molecular Biology Medical Genetics and Genomics Medical Biotechnology (Focus on Cell Biology, (incl. Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:kth:diva-366187 (URN)10.1186/s13059-025-03616-4 (DOI)001502167900001 ()40462185 (PubMedID)2-s2.0-105007441526 (Scopus ID)
Note

Not duplicate with DiVA 1959447

QC 20250707

Available from: 2025-07-07 Created: 2025-07-07 Last updated: 2025-08-15Bibliographically approved
Song, X., Jin, H., Li, X., Yuan, M., Yang, H., Sato, Y., . . . Mardinoglu, A. (2025). Systematically identification of survival-associated eQTLs in a Japanese kidney cancer cohort. PLOS Genetics, 21(7 July), Article ID e1011770.
Open this publication in new window or tab >>Systematically identification of survival-associated eQTLs in a Japanese kidney cancer cohort
Show others...
2025 (English)In: PLOS Genetics, ISSN 1553-7390, E-ISSN 1553-7404, Vol. 21, no 7 July, article id e1011770Article in journal (Refereed) Published
Abstract [en]

Background Clear cell renal carcinoma (ccRCC) is the predominant form of kidney cancer, but the prognostic value of expression quantitative trait loci (eQTLs) remains underexplored, particularly in Asian populations. Objective We analyzed whole-exome sequencing and RNA sequencing data from 100 Japanese ccRCC patients to identify eQTLs. Multiple Cox proportional hazard models assessed survival associations, with validation in the Cancer Genome Atlas ccRCC cohort (n = 287). Results We identified 805 eGenes and 4,558 cis-eQTLs in the Japanese cohort. Survival analysis revealed a total of 9 eGenes significantly associated with overall survival (FDR < 0.05). Further exploratory analysis were performed using 158 eGenes and 711 eQTLs (p-value <0.05) as potential prognostic signals. Among these, 223 eQTLs regulating 54 eGenes showed consistent prognostic effects at both expression and genetic levels. Cross-population validation identified eight eQTLs regulating 11 eGenes with reproducible survival associations across ethnicities, including a missense mutation in ERV3–1 and regulatory variants near ANKRD20A7P. These variants demonstrated consistent allelic effects on both gene expression and patient survival in both cohorts.

Place, publisher, year, edition, pages
Public Library of Science (PLoS), 2025
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:kth:diva-368942 (URN)10.1371/journal.pgen.1011770 (DOI)001524169900006 ()40622919 (PubMedID)2-s2.0-105009893848 (Scopus ID)
Note

QC 20250828

Available from: 2025-08-28 Created: 2025-08-28 Last updated: 2025-10-10Bibliographically approved
Kaynar, A., Ozcan, M., Li, X., Turkez, H., Zhang, C., Uhlén, M., . . . Mardinoglu, A. (2024). Discovery of a Therapeutic Agent for Glioblastoma Using a Systems Biology-Based Drug Repositioning Approach. International Journal of Molecular Sciences, 25(14), Article ID 7868.
Open this publication in new window or tab >>Discovery of a Therapeutic Agent for Glioblastoma Using a Systems Biology-Based Drug Repositioning Approach
Show others...
2024 (English)In: International Journal of Molecular Sciences, ISSN 1661-6596, E-ISSN 1422-0067, Vol. 25, no 14, article id 7868Article in journal (Refereed) Published
Abstract [en]

Glioblastoma (GBM), a highly malignant tumour of the central nervous system, presents with a dire prognosis and low survival rates. The heterogeneous and recurrent nature of GBM renders current treatments relatively ineffective. In our study, we utilized an integrative systems biology approach to uncover the molecular mechanisms driving GBM progression and identify viable therapeutic drug targets for developing more effective GBM treatment strategies. Our integrative analysis revealed an elevated expression of CHST2 in GBM tumours, designating it as an unfavourable prognostic gene in GBM, as supported by data from two independent GBM cohorts. Further, we pinpointed WZ-4002 as a potential drug candidate to modulate CHST2 through computational drug repositioning. WZ-4002 directly targeted EGFR (ERBB1) and ERBB2, affecting their dimerization and influencing the activity of adjacent genes, including CHST2. We validated our findings by treating U-138 MG cells with WZ-4002, observing a decrease in CHST2 protein levels and a reduction in cell viability. In summary, our research suggests that the WZ-4002 drug candidate may effectively modulate CHST2 and adjacent genes, offering a promising avenue for developing efficient treatment strategies for GBM patients.

Place, publisher, year, edition, pages
Multidisciplinary Digital Publishing Institute (MDPI), 2024
Keywords
co-expression, drug repositioning, extracellular matrix, glioblastoma, glycosaminoglycans, survival
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:kth:diva-351705 (URN)10.3390/ijms25147868 (DOI)001277288800001 ()2-s2.0-85199800918 (Scopus ID)
Note

QC 20240814

Available from: 2024-08-13 Created: 2024-08-13 Last updated: 2024-08-14Bibliographically approved
Jin, H., Kim, W., Yuan, M., Li, X., Yang, H., Li, M., . . . Mardinoglu, A. (2024). Identification of SPP1+ macrophages as an immune suppressor in hepatocellular carcinoma using single-cell and bulk transcriptomics. Frontiers in Immunology, 15, Article ID 1446453.
Open this publication in new window or tab >>Identification of SPP1+ macrophages as an immune suppressor in hepatocellular carcinoma using single-cell and bulk transcriptomics
Show others...
2024 (English)In: Frontiers in Immunology, E-ISSN 1664-3224, Vol. 15, article id 1446453Article in journal (Refereed) Published
Abstract [en]

Introduction: Macrophages and T cells play crucial roles in liver physiology, but their functional diversity in hepatocellular carcinoma (HCC) remains largely unknown. Methods: Two bulk RNA-sequencing (RNA-seq) cohorts for HCC were analyzed using gene co-expression network analysis. Key gene modules and networks were mapped to single-cell RNA-sequencing (scRNA-seq) data of HCC. Cell type fraction of bulk RNA-seq data was estimated by deconvolution approach using single-cell RNA-sequencing data as a reference. Survival analysis was carried out to estimate the prognosis of different immune cell types in bulk RNA-seq cohorts. Cell-cell interaction analysis was performed to identify potential links between immune cell types in HCC. Results: In this study, we analyzed RNA-seq data from two large-scale HCC cohorts, revealing a major and consensus gene co-expression cluster with significant implications for immunosuppression. Notably, these genes exhibited higher enrichment in liver macrophages than T cells, as confirmed by scRNA-seq data from HCC patients. Integrative analysis of bulk and single-cell RNA-seq data pinpointed SPP1+ macrophages as an unfavorable cell type, while VCAN+ macrophages, C1QA+ macrophages, and CD8+ T cells were associated with a more favorable prognosis for HCC patients. Subsequent scRNA-seq investigations and in vitro experiments elucidated that SPP1, predominantly secreted by SPP1+ macrophages, inhibits CD8+ T cell proliferation. Finally, targeting SPP1 in tumor-associated macrophages through inhibition led to a shift towards a favorable phenotype. Discussion: This study underpins the potential of SPP1 as a translational target in immunotherapy for HCC.

Place, publisher, year, edition, pages
Frontiers Media SA, 2024
Keywords
co-expression network, hepatocellular carcinoma, macrophage heterogeneity, single-cell sequencing, tumor-associated macrophage
National Category
Cancer and Oncology Cell and Molecular Biology
Identifiers
urn:nbn:se:kth:diva-358179 (URN)10.3389/fimmu.2024.1446453 (DOI)001378522100001 ()39691723 (PubMedID)2-s2.0-85212417452 (Scopus ID)
Note

QC 20250116

Available from: 2025-01-07 Created: 2025-01-07 Last updated: 2025-05-07Bibliographically approved
Meng, L., Jin, H., Yulug, B., Altay, Ö., Li, X., Hanoglu, L., . . . Mardinoglu, A. (2024). Multi-omics analysis reveals the key factors involved in the severity of the Alzheimer's disease. Alzheimer's Research & Therapy, 16(1), Article ID 213.
Open this publication in new window or tab >>Multi-omics analysis reveals the key factors involved in the severity of the Alzheimer's disease
Show others...
2024 (English)In: Alzheimer's Research & Therapy, E-ISSN 1758-9193, Vol. 16, no 1, article id 213Article in journal (Refereed) Published
Abstract [en]

Alzheimer's disease (AD) is a debilitating neurodegenerative disorder with a global impact, yet its pathogenesis remains poorly understood. While age, metabolic abnormalities, and accumulation of neurotoxic substances are potential risk factors for AD, their effects are confounded by other factors. To address this challenge, we first utilized multi-omics data from 87 well phenotyped AD patients and generated plasma proteomics and metabolomics data, as well as gut and saliva metagenomics data to investigate the molecular-level alterations accounting the host-microbiome interactions. Second, we analyzed individual omics data and identified the key parameters involved in the severity of the dementia in AD patients. Next, we employed Artificial Intelligence (AI) based models to predict AD severity based on the significantly altered features identified in each omics analysis. Based on our integrative analysis, we found the clinical relevance of plasma proteins, including SKAP1 and NEFL, plasma metabolites including homovanillate and glutamate, and Paraprevotella clara in gut microbiome in predicting the AD severity. Finally, we validated the predictive power of our AI based models by generating additional multi-omics data from the same group of AD patients by following up for 3 months. Hence, we observed that these results may have important implications for the development of potential diagnostic and therapeutic approaches for AD patients.

Place, publisher, year, edition, pages
Springer Nature, 2024
National Category
Bioinformatics and Computational Biology
Identifiers
urn:nbn:se:kth:diva-355155 (URN)10.1186/s13195-024-01578-6 (DOI)001327038600001 ()39358810 (PubMedID)2-s2.0-85205527457 (Scopus ID)
Note

QC 20241024

Available from: 2024-10-24 Created: 2024-10-24 Last updated: 2025-02-07Bibliographically approved
Ceyhan, A. B., Ozcan, M., Kim, W., Li, X., Altay, Ö., Zhang, C. & Mardinoglu, A. (2024). Novel drug targets and molecular mechanisms for sarcopenia based on systems biology. Biomedicine and Pharmacotherapy, 176, Article ID 116920.
Open this publication in new window or tab >>Novel drug targets and molecular mechanisms for sarcopenia based on systems biology
Show others...
2024 (English)In: Biomedicine and Pharmacotherapy, ISSN 0753-3322, E-ISSN 1950-6007, Vol. 176, article id 116920Article in journal (Refereed) Published
Abstract [en]

Sarcopenia is a major public health concern among older adults, leading to disabilities, falls, fractures, and mortality. This study aimed to elucidate the pathophysiological mechanisms of sarcopenia and identify potential therapeutic targets using systems biology approaches. RNA-seq data from muscle biopsies of 24 sarcopenic and 29 healthy individuals from a previous cohort were analysed. Differential expression, gene set enrichment, gene co-expression network, and topology analyses were conducted to identify target genes implicated in sarcopenia pathogenesis, resulting in the selection of 6 hub genes (PDHX, AGL, SEMA6C, CASQ1, MYORG, and CCDC69). A drug repurposing approach was then employed to identify new pharmacological treatment options for sarcopenia (clofibric-acid, troglitazone, withaferin-a, palbociclib, MG-132, bortezomib). Finally, validation experiments in muscle cell line (C2C12) revealed MG-132 and troglitazone as promising candidates for sarcopenia treatment. Our approach, based on systems biology and drug repositioning, provides insight into the molecular mechanisms of sarcopenia and offers potential new treatment options using existing drugs.

Place, publisher, year, edition, pages
Elsevier BV, 2024
Keywords
Co-expression network analysis, Differential expression analysis, Drug repurposing, Sarcopenia, System biology, Translational medicine
National Category
Geriatrics
Identifiers
urn:nbn:se:kth:diva-348306 (URN)10.1016/j.biopha.2024.116920 (DOI)001253736800001 ()38876054 (PubMedID)2-s2.0-85195638221 (Scopus ID)
Note

QC 20240624

Available from: 2024-06-20 Created: 2024-06-20 Last updated: 2024-07-05Bibliographically approved
Yulug, B., Altay, Ö., Li, X., Hanoglu, L., Cankaya, S., Lam, S., . . . Mardinoglu, A. (2023). Combined metabolic activators improve cognitive functions in Alzheimer's disease patients: a randomised, double-blinded, placebo-controlled phase-II trial. Translational Neurodegeneration, 12(1), Article ID 4.
Open this publication in new window or tab >>Combined metabolic activators improve cognitive functions in Alzheimer's disease patients: a randomised, double-blinded, placebo-controlled phase-II trial
Show others...
2023 (English)In: Translational Neurodegeneration, ISSN 2047-9158, Vol. 12, no 1, article id 4Article in journal (Refereed) Published
Abstract [en]

Background Alzheimer's disease (AD) is associated with metabolic abnormalities linked to critical elements of neurodegeneration. We recently administered combined metabolic activators (CMA) to the AD rat model and observed that CMA improves the AD-associated histological parameters in the animals. CMA promotes mitochondrial fatty acid uptake from the cytosol, facilitates fatty acid oxidation in the mitochondria, and alleviates oxidative stress.Methods Here, we designed a randomised, double-blinded, placebo-controlled phase-II clinical trial and studied the effect of CMA administration on the global metabolism of AD patients. One-dose CMA included 12.35 g L-serine (61.75%), 1 g nicotinamide riboside (5%), 2.55 g N-acetyl-L-cysteine (12.75%), and 3.73 g L-carnitine tartrate (18.65%). AD patients received one dose of CMA or placebo daily during the first 28 days and twice daily between day 28 and day 84. The primary endpoint was the difference in the cognitive function and daily living activity scores between the placebo and the treatment arms. The secondary aim of this study was to evaluate the safety and tolerability of CMA. A comprehensive plasma metabolome and proteome analysis was also performed to evaluate the efficacy of the CMA in AD patients.Results We showed a significant decrease of AD Assessment Scale-cognitive subscale (ADAS-Cog) score on day 84 vs day 0 (P = 0.00001, 29% improvement) in the CMA group. Moreover, there was a significant decline (P = 0.0073) in ADAS-Cog scores (improvement of cognitive functions) in the CMA compared to the placebo group in patients with higher ADAS-Cog scores. Improved cognitive functions in AD patients were supported by the relevant alterations in the hippocampal volumes and cortical thickness based on imaging analysis. Moreover, the plasma levels of proteins and metabolites associated with NAD + and glutathione metabolism were significantly improved after CMA treatment.Conclusion Our results indicate that treatment of AD patients with CMA can lead to enhanced cognitive functions and improved clinical parameters associated with phenomics, metabolomics, proteomics and imaging analysis.

Place, publisher, year, edition, pages
Springer Nature, 2023
Keywords
Alzheimer's disease, Combined metabolic activators, Multi-omics, Systems biology, Systems medicine
National Category
Neurology
Identifiers
urn:nbn:se:kth:diva-324396 (URN)10.1186/s40035-023-00336-2 (DOI)000922978700001 ()36703196 (PubMedID)2-s2.0-85146871450 (Scopus ID)
Note

QC 20230301

Available from: 2023-03-01 Created: 2023-03-01 Last updated: 2023-12-07Bibliographically approved
Graves, O. K., Kim, W., Ozcan, M., Ashraf, S., Turkez, H., Yuan, M., . . . Li, X. (2023). Discovery of drug targets and therapeutic agents based on drug repositioning to treat lung adenocarcinoma. Biomedicine and Pharmacotherapy, 161, Article ID 114486.
Open this publication in new window or tab >>Discovery of drug targets and therapeutic agents based on drug repositioning to treat lung adenocarcinoma
Show others...
2023 (English)In: Biomedicine and Pharmacotherapy, ISSN 0753-3322, E-ISSN 1950-6007, Vol. 161, article id 114486Article in journal (Refereed) Published
Abstract [en]

Background: Lung adenocarcinoma (LUAD) is the one of the most common subtypes in lung cancer. Although various targeted therapies have been used in the clinical practice, the 5-year overall survival rate of patients is still low. Thus, it is urgent to identify new therapeutic targets and develop new drugs for the treatment of the LUAD patients. Methods: Survival analysis was used to identify the prognostic genes. Gene co-expression network analysis was used to identify the hub genes driving the tumor development. A profile-based drug repositioning approach was used to repurpose the potentially useful drugs for targeting the hub genes. MTT and LDH assay were used to measure the cell viability and drug cytotoxicity, respectively. Western blot was used to detect the expression of the proteins. Findings: We identified 341 consistent prognostic genes from two independent LUAD cohorts, whose high expression was associated with poor survival outcomes of patients. Among them, eight genes were identified as hub genes due to their high centrality in the key functional modules in the gene-co-expression network analysis and these genes were associated with the various hallmarks of cancer (e.g., DNA replication and cell cycle). We performed drug repositioning analysis for three of the eight genes (CDCA8, MCM6, and TTK) based on our drug repositioning approach. Finally, we repurposed five drugs for inhibiting the protein expression level of each target gene and validated the drug efficacy by performing in vitro experiments. Interpretation: We found the consensus targetable genes for the treatment of LUAD patients with different races and geographic characteristics. We also proved the feasibility of our drug repositioning approach for the development of new drugs for disease treatment.

Place, publisher, year, edition, pages
Elsevier BV, 2023
Keywords
Lung adenocarcinoma, Co-expression network, Target identification, Drug repositioning
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:kth:diva-326058 (URN)10.1016/j.biopha.2023.114486 (DOI)000955057800001 ()36906970 (PubMedID)2-s2.0-85149805666 (Scopus ID)
Note

QC 20230425

Available from: 2023-04-25 Created: 2023-04-25 Last updated: 2023-04-25Bibliographically approved
Bayraktar, A., Li, X., Kim, W., Zhang, C., Turkez, H., Shoaie, S. & Mardinoglu, A. (2023). Drug repositioning targeting glutaminase reveals drug candidates for the treatment of Alzheimer's disease patients. Journal of Translational Medicine, 21(1), Article ID 332.
Open this publication in new window or tab >>Drug repositioning targeting glutaminase reveals drug candidates for the treatment of Alzheimer's disease patients
Show others...
2023 (English)In: Journal of Translational Medicine, E-ISSN 1479-5876, Vol. 21, no 1, article id 332Article in journal (Refereed) Published
Abstract [en]

Background

Despite numerous clinical trials and decades of endeavour, there is still no effective cure for Alzheimer's disease. Computational drug repositioning approaches may be employed for the development of new treatment strategies for Alzheimer's patients since an extensive amount of omics data has been generated during pre-clinical and clinical studies. However, targeting the most critical pathophysiological mechanisms and determining drugs with proper pharmacodynamics and good efficacy are equally crucial in drug repurposing and often imbalanced in Alzheimer's studies.

Methods

Here, we investigated central co-expressed genes upregulated in Alzheimer's disease to determine a proper therapeutic target. We backed our reasoning by checking the target gene's estimated non-essentiality for survival in multiple human tissues. We screened transcriptome profiles of various human cell lines perturbed by drug induction (for 6798 compounds) and gene knockout using data available in the Connectivity Map database. Then, we applied a profile-based drug repositioning approach to discover drugs targeting the target gene based on the correlations between these transcriptome profiles. We evaluated the bioavailability, functional enrichment profiles and drug-protein interactions of these repurposed agents and evidenced their cellular viability and efficacy in glial cell culture by experimental assays and Western blotting. Finally, we evaluated their pharmacokinetics to anticipate to which degree their efficacy can be improved.

Results

We identified glutaminase as a promising drug target. Glutaminase overexpression may fuel the glutamate excitotoxicity in neurons, leading to mitochondrial dysfunction and other neurodegeneration hallmark processes. The computational drug repurposing revealed eight drugs: mitoxantrone, bortezomib, parbendazole, crizotinib, withaferin-a, SA-25547 and two unstudied compounds. We demonstrated that the proposed drugs could effectively suppress glutaminase and reduce glutamate production in the diseased brain through multiple neurodegeneration-associated mechanisms, including cytoskeleton and proteostasis. We also estimated the human blood-brain barrier permeability of parbendazole and SA-25547 using the SwissADME tool.

Conclusions

This study method effectively identified an Alzheimer's disease marker and compounds targeting the marker and interconnected biological processes by use of multiple computational approaches. Our results highlight the importance of synaptic glutamate signalling in Alzheimer's disease progression. We suggest repurposable drugs (like parbendazole) with well-evidenced activities that we linked to glutamate synthesis hereby and novel molecules (SA-25547) with estimated mechanisms for the treatment of Alzheimer's patients.

Place, publisher, year, edition, pages
BMC, 2023
Keywords
Alzheimer's disease, Profile-based computational drug repositioning, Glutaminase, Anti-carcinogenic drugs, Gene co-expression network analysis, Parbendazole
National Category
Neurosciences Pharmaceutical Sciences
Identifiers
urn:nbn:se:kth:diva-328435 (URN)10.1186/s12967-023-04192-6 (DOI)000993072700003 ()37210557 (PubMedID)2-s2.0-85159698366 (Scopus ID)
Note

QC 20230612

Available from: 2023-06-12 Created: 2023-06-12 Last updated: 2024-07-04Bibliographically approved
Yang, H., Li, X., Jin, H., Turkez, H., Ozturk, G., Doganay, H. L., . . . Mardinoglu, A. (2023). Longitudinal metabolomics analysis reveals the acute effect of cysteine and NAC included in the combined metabolic activators. Free Radical Biology & Medicine, 204, 347-358
Open this publication in new window or tab >>Longitudinal metabolomics analysis reveals the acute effect of cysteine and NAC included in the combined metabolic activators
Show others...
2023 (English)In: Free Radical Biology & Medicine, ISSN 0891-5849, E-ISSN 1873-4596, Vol. 204, p. 347-358Article in journal (Refereed) Published
Abstract [en]

Growing evidence suggests that the depletion of plasma NAD+ and glutathione (GSH) may play an important role in the development of metabolic disorders. The administration of Combined Metabolic Activators (CMA), consisting of GSH and NAD+ precursors, has been explored as a promising therapeutic strategy to target multiple altered pathways associated with the pathogenesis of the diseases. Although studies have examined the therapeutic effect of CMA that contains N-acetyl-L-cysteine (NAC) as a metabolic activator, a system-wide comparison of the metabolic response to the administration of CMA with NAC and cysteine remains lacking. In this placebo-controlled study, we studied the acute effect of the CMA administration with different metabolic activators, including NAC or cysteine with/without nicotinamide or flush free niacin, and performed longitudinal untargeted-metabolomics profiling of plasma obtained from 70 well-characterized healthy volunteers. The time-series metabolomics data revealed the metabolic pathways affected after the administration of CMAs showed high similarity between CMA containing nicotinamide and NAC or cysteine as metabolic activators. Our analysis also showed that CMA with cysteine is well-tolerated and safe in healthy individuals throughout the study. Last, our study systematically provided insights into a complex and dynamics landscape involved in amino acid, lipid and nicotinamide metabolism, reflecting the metabolic responses to CMA administration containing different metabolic activators.

Place, publisher, year, edition, pages
Elsevier BV, 2023
Keywords
Combined metabolic activators, Cysteine, Metabolomics, N-Acetyl-L-cysteine, Niacin, Nicotinamide
National Category
Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:kth:diva-331535 (URN)10.1016/j.freeradbiomed.2023.05.013 (DOI)001014625400001 ()37245532 (PubMedID)2-s2.0-85160423966 (Scopus ID)
Note

QC 20230711

Available from: 2023-07-11 Created: 2023-07-11 Last updated: 2024-05-16Bibliographically approved
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0002-8301-9959

Search in DiVA

Show all publications