kth.sePublications
System disruptions
We are currently experiencing disruptions on the search portals due to high traffic. We are working to resolve the issue, you may temporarily encounter an error message.
Change search
Link to record
Permanent link

Direct link
Publications (10 of 111) Show all publications
Yang, H., Atak, D., Yuan, M., Li, M., Altay, Ö., Demirtas, E., . . . Zeybel, M. (2025). Integrative proteo-transcriptomic characterization of advanced fibrosis in chronic liver disease across etiologies. Cell Reports Medicine, 6(2), Article ID 101935.
Open this publication in new window or tab >>Integrative proteo-transcriptomic characterization of advanced fibrosis in chronic liver disease across etiologies
Show others...
2025 (English)In: Cell Reports Medicine, E-ISSN 2666-3791, Vol. 6, no 2, article id 101935Article in journal (Refereed) Published
Abstract [en]

Chronic hepatic injury and inflammation from various causes can lead to fibrosis and cirrhosis, potentially predisposing to hepatocellular carcinoma. The molecular mechanisms underlying fibrosis and its progression remain incompletely understood. Using a proteo-transcriptomics approach, we analyze liver and plasma samples from 330 individuals, including 40 healthy individuals and 290 patients with histologically characterized fibrosis due to chronic viral infection, alcohol consumption, or metabolic dysfunction-associated steatotic liver disease. Our findings reveal dysregulated pathways related to extracellular matrix, immune response, inflammation, and metabolism in advanced fibrosis. We also identify 132 circulating proteins associated with advanced fibrosis, with neurofascin and growth differentiation factor 15 demonstrating superior predictive performance for advanced fibrosis(area under the receiver operating characteristic curve [AUROC] 0.89 [95% confidence interval (CI) 0.81–0.97]) compared to the fibrosis-4 model (AUROC 0.85 [95% CI 0.78–0.93]). These findings provide insights into fibrosis pathogenesis and highlight the potential for more accurate non-invasive diagnosis.

Place, publisher, year, edition, pages
Elsevier BV, 2025
Keywords
chronic liver disease, liver fibrosis, multi-omics, non-invasive, systems biology
National Category
Gastroenterology and Hepatology
Identifiers
urn:nbn:se:kth:diva-360591 (URN)10.1016/j.xcrm.2025.101935 (DOI)001434169900001 ()39889710 (PubMedID)2-s2.0-85217935601 (Scopus ID)
Note

QC 20250318

Available from: 2025-02-26 Created: 2025-02-26 Last updated: 2025-03-18Bibliographically approved
Yulug, B., Altay, Ö., Lei, X., Hanoglu, L., Cankaya, S., Velioglu, H. A., . . . Mardinoglu, A. (2025). Multi-omics characterization of improved cognitive functions in Parkinson’s disease patients after the combined metabolic activator treatment: a randomized, double-blinded, placebo-controlled phase II trial. Brain Communications, 7(1), Article ID fcae478.
Open this publication in new window or tab >>Multi-omics characterization of improved cognitive functions in Parkinson’s disease patients after the combined metabolic activator treatment: a randomized, double-blinded, placebo-controlled phase II trial
Show others...
2025 (English)In: Brain Communications, E-ISSN 2632-1297, Vol. 7, no 1, article id fcae478Article in journal (Refereed) Published
Abstract [en]

Parkinson’s disease is primarily marked by mitochondrial dysfunction and metabolic abnormalities. We recently reported that the combined metabolic activators improved the immunohistochemical parameters and behavioural functions in Parkinson’s disease and Alzheimer’s disease animal models and the cognitive functions in Alzheimer’s disease patients. These metabolic activators serve as the precursors of nicotinamide adenine dinucleotide and glutathione, and they can be used to activate mitochondrial metabolism and eventually treat mitochondrial dysfunction. Here, we designed a randomized, double-blinded, placebo-controlled phase II study in Parkinson’s disease patients with 84 days combined metabolic activator administration. A single dose of combined metabolic activator contains L-serine (12.35 g), N-acetyl-L-cysteine (2.55 g), nicotinamide riboside (1 g) and L-carnitine tartrate (3.73 g). Patients were administered either one dose of combined metabolic activator or a placebo daily for the initial 28 days, followed by twice-daily dosing for the next 56 days. The main goal of the study was to evaluate the clinical impact on motor functions using the Unified Parkinson’s Disease Rating Scale and to determine the safety and tolerability of combined metabolic activator. A secondary objective was to assess cognitive functions utilizing the Montreal Cognitive Assessment and to analyse brain activity through functional MRI. We also performed comprehensive plasma metabolomics and proteomics analysis for detailed characterization of Parkinson’s disease patients who participated in the study. Although no improvement in motor functions was observed, cognitive function was shown to be significantly improved (P < 0.0000) in Parkinson’s disease patients treated with the combined metabolic activator group over 84 days, whereas no such improvement was noted in the placebo group (P > 0.05). Moreover, a significant reduction (P = 0.001) in Montreal Cognitive Assessment scores was observed in the combined metabolic activator group, with no decline (P > 0.05) in the placebo group among severe Parkinson’s disease patients with lower baseline Montreal Cognitive Assessment scores. We showed that improvement in cognition was associated with critical brain network alterations based on functional MRI analysis, especially relevant to areas with cognitive functions in the brain. Finally, through a comprehensive multi-omics analysis, we elucidated the molecular mechanisms underlying cognitive improvements observed in Parkinson’s disease patients. Our results show that combined metabolic activator administration leads to enhanced cognitive function and improved metabolic health in Parkinson’s disease patients as recently shown in Alzheimer’s disease patients.

Place, publisher, year, edition, pages
Oxford University Press (OUP), 2025
Keywords
combined metabolic activators, multi-omics, Parkinson’s disease, systems biology
National Category
Neurosciences Neurology
Identifiers
urn:nbn:se:kth:diva-359300 (URN)10.1093/braincomms/fcae478 (DOI)001397642700001 ()2-s2.0-85215432829 (Scopus ID)
Note

QC 20250131

Available from: 2025-01-29 Created: 2025-01-29 Last updated: 2025-02-13Bibliographically approved
Yang, H., Zhang, C., Kim, W., Shi, M., Kiliclioglu, M., Bayram, C., . . . Mardinoglu, A. (2025). Multi-tissue network analysis reveals the effect of JNK inhibition on dietary sucrose-induced metabolic dysfunction in rats. eLIFE, 13, Article ID RP98427.
Open this publication in new window or tab >>Multi-tissue network analysis reveals the effect of JNK inhibition on dietary sucrose-induced metabolic dysfunction in rats
Show others...
2025 (English)In: eLIFE, E-ISSN 2050-084X, Vol. 13, article id RP98427Article in journal (Refereed) Published
Abstract [en]

Excessive consumption of sucrose, in the form of sugar-sweetened beverages, has been implicated in the pathogenesis of metabolic dysfunction-associated fatty liver disease (MAFLD) and other related metabolic syndromes. The c-Jun N-terminal kinase (JNK) pathway plays a crucial role in response to dietary stressors, and it was demonstrated that the inhibition of the JNK pathway could potentially be used in the treatment of MAFLD. However, the intricate mechanisms underlying these interventions remain incompletely understood given their multifaceted effects across multiple tissues. In this study, we challenged rats with sucrose-sweetened water and investigated the potential effects of JNK inhibition by employing network analysis based on the transcriptome profiling obtained from hepatic and extrahepatic tissues, including visceral white adipose tissue, skeletal muscle, and brain. Our data demonstrate that JNK inhibition by JNK-IN-5A effectively reduces the circulating triglyceride accumulation and inflammation in rats subjected to sucrose consumption. Coexpression analysis and genome-scale metabolic modeling reveal that sucrose overconsumption primarily induces transcriptional dysfunction related to fatty acid and oxidative metabolism in the liver and adipose tissues, which are largely rectified after JNK inhibition at a clinically relevant dose. Skeletal muscle exhibited minimal transcriptional changes to sucrose overconsumption but underwent substantial metabolic adaptation following the JNK inhibition. Overall, our data provides novel insights into the molecular basis by which JNK inhibition exerts its metabolic effect in the metabolically active tissues. Furthermore, our findings underpin the critical role of extrahepatic metabolism in the development of diet-induced steatosis, offering valuable guidance for future studies focused on JNK-targeting for effective treatment of MAFLD.

Place, publisher, year, edition, pages
eLife Sciences Publications, Ltd, 2025
Keywords
MAFLD, JNK, sucrose, JNK-IN-5A, multi-tissue transcriptome, Rat
National Category
Basic Medicine
Identifiers
urn:nbn:se:kth:diva-360435 (URN)10.7554/eLife.98427 (DOI)001420073300001 ()39932177 (PubMedID)2-s2.0-85218435359 (Scopus ID)
Note

QC 20250303

Available from: 2025-02-26 Created: 2025-02-26 Last updated: 2025-03-03Bibliographically approved
Sebhaoui, J., Ashraf, S., Iqbal, S., Hajji, M., Belmen, B., Yesilyurt, G., . . . Mardinoglu, A. (2025). Synthesis of novel tetrahydrobenzo[b]thiophene-3-carbonitrile (THBTC)-based heterocycles: Structural insights, reactivity profiles, and in-silico bioactivity studies. Journal of Molecular Structure, 1326, Article ID 141110.
Open this publication in new window or tab >>Synthesis of novel tetrahydrobenzo[b]thiophene-3-carbonitrile (THBTC)-based heterocycles: Structural insights, reactivity profiles, and in-silico bioactivity studies
Show others...
2025 (English)In: Journal of Molecular Structure, ISSN 0022-2860, E-ISSN 1872-8014, Vol. 1326, article id 141110Article in journal (Refereed) Published
Abstract [en]

This study reports the synthesis and characterization of novel 4,5,6,7-tetrahydrobenzo[b]thiophene-3-carbonitrile (THBTC) derivatives. Compounds 3–12 were synthesized via N-alkylation of compound 2 with various alkyl, benzyl, and heterocyclic halides. Notably, compound 11 was formed through an unexpected intramolecular cyclization mechanism. Furthermore, derivatives 14–24, incorporating a 1,2,3-triazole ring, were prepared using a Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) reaction between compound 12 and readily synthesized azides. All derivatives were characterized by ¹H NMR, ¹³C NMR, and mass spectrometry. Single-crystal X-ray diffraction analysis of derivatives 11 and 25 confirmed their molecular structures and revealed the presence of intramolecular cyclization and tautomerism. The crystal arrangements exhibited a range of noncovalent interactions, including N—H···O, C—H···O, N—H···π, and π-π stacking, which contributed to their stability in the solid state. A computational study using QTAIM and IGM topological analyses was conducted, offering insights into the nature and strength of intermolecular noncovalent interactions. Additionally, conceptual DFT calculations at wB97X-D/cc-pVTZ level provided insights into the global and local reactivity properties of both compounds. Molecular docking studies were conducted to evaluate their binding characteristics as inhibitors of the JNK3 target protein. Finally, in silico ADME (absorption, distribution, metabolism, and excretion) predictions were performed to assess their druglikeness and bioavailability. This work aims to advance our understanding of the chemistry, and potential applications of 4,5,6,7-tetrahydrobenzo[b]thiophene-3-carbonitrile (THBTC) derivatives.

Place, publisher, year, edition, pages
Elsevier BV, 2025
Keywords
4 5 6 7-Tetrahydrobenzo[b]thiophene-3, carbonitrile derivatives, N-alkylation/CuAAC synthesis, X-ray crystallography, DFT calculations, Molecular docking, ADME predictions
National Category
Organic Chemistry
Identifiers
urn:nbn:se:kth:diva-359499 (URN)10.1016/j.molstruc.2024.141110 (DOI)001381318800001 ()2-s2.0-85212573920 (Scopus ID)
Note

QC 20250204

Available from: 2025-02-04 Created: 2025-02-04 Last updated: 2025-02-04Bibliographically approved
Yuan, M., Zhang, C., von Feilitzen, K., Zwahlen, M., Shi, M., Li, X., . . . Mardinoglu, A. (2025). The Human Pathology Atlas for deciphering the prognostic features of human cancers. EBioMedicine, 111, Article ID 105495.
Open this publication in new window or tab >>The Human Pathology Atlas for deciphering the prognostic features of human cancers
Show others...
2025 (English)In: EBioMedicine, E-ISSN 2352-3964, Vol. 111, article id 105495Article in journal (Refereed) Published
Abstract [en]

Background: Cancer is one of the leading causes of mortality worldwide, highlighting the urgent need for a deeper molecular understanding and the development of personalized treatments. The present study aims to establish a solid association between gene expression and patient survival outcomes to enhance the utility of the Human Pathology Atlas for cancer research. Methods: In this updated analysis, we examined the expression profiles of 6918 patients across 21 cancer types. We integrated data from 10 independent cancer cohorts, creating a cross-validated, reliable collection of prognostic genes. We applied systems biology approach to identify the association between gene expression profiles and patient survival outcomes. We further constructed prognostic regulatory networks for kidney renal clear cell carcinoma (KIRC) and liver hepatocellular carcinoma (LIHC), which elucidate the molecular underpinnings associated with patient survival in these cancers. Findings: We observed that gene expression during the transition from normal to tumorous tissue exhibited diverse shifting patterns in their original tissue locations. Significant correlations between gene expression and patient survival outcomes were identified in KIRC and LIHC among the major cancer types. Additionally, the prognostic regulatory network established for these two cancers showed the indicative capabilities of the Human Pathology Atlas and provides actionable insights for cancer research. Interpretation: The updated Human Pathology Atlas provides a significant foundation for precision oncology and the formulation of personalized treatment strategies. These findings deepen our understanding of cancer biology and have the potential to advance targeted therapeutic approaches in clinical practice. Funding: The Knut and Alice Wallenberg Foundation ( 72110), the China Scholarship Council (Grant No. 202006940003).

Place, publisher, year, edition, pages
Elsevier BV, 2025
Keywords
Cancer, Survival, Systems biology, Transcriptomics
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:kth:diva-357900 (URN)10.1016/j.ebiom.2024.105495 (DOI)001425050600001 ()39662180 (PubMedID)2-s2.0-85211197830 (Scopus ID)
Note

QC 20250303

Available from: 2024-12-19 Created: 2024-12-19 Last updated: 2025-03-03Bibliographically approved
Altay, Ö., Yang, H., Yildirim, S., Bayram, C., Bolat, I., Oner, S., . . . Mardinoglu, A. (2024). Combined Metabolic Activators with Different NAD+ Precursors Improve Metabolic Functions in the Animal Models of Neurodegenerative Diseases. Biomedicines, 12(4), Article ID 927.
Open this publication in new window or tab >>Combined Metabolic Activators with Different NAD+ Precursors Improve Metabolic Functions in the Animal Models of Neurodegenerative Diseases
Show others...
2024 (English)In: Biomedicines, E-ISSN 2227-9059, Vol. 12, no 4, article id 927Article in journal (Refereed) Published
Abstract [en]

Background: Mitochondrial dysfunction and metabolic abnormalities are acknowledged as significant factors in the onset of neurodegenerative disorders such as Parkinson’s disease (PD) and Alzheimer’s disease (AD). Our research has demonstrated that the use of combined metabolic activators (CMA) may alleviate metabolic dysfunctions and stimulate mitochondrial metabolism. Therefore, the use of CMA could potentially be an effective therapeutic strategy to slow down or halt the progression of PD and AD. CMAs include substances such as the glutathione precursors (L-serine and N-acetyl cysteine), the NAD+ precursor (nicotinamide riboside), and L-carnitine tartrate. Methods: Here, we tested the effect of two different formulations, including CMA1 (nicotinamide riboside, L-serine, N-acetyl cysteine, L-carnitine tartrate), and CMA2 (nicotinamide, L-serine, N-acetyl cysteine, L-carnitine tartrate), as well as their individual components, on the animal models of AD and PD. We assessed the brain and liver tissues for pathological changes and immunohistochemical markers. Additionally, in the case of PD, we performed behavioral tests and measured responses to apomorphine-induced rotations. Findings: Histological analysis showed that the administration of both CMA1 and CMA2 formulations led to improvements in hyperemia, degeneration, and necrosis in neurons for both AD and PD models. Moreover, the administration of CMA2 showed a superior effect compared to CMA1. This was further corroborated by immunohistochemical data, which indicated a reduction in immunoreactivity in the neurons. Additionally, notable metabolic enhancements in liver tissues were observed using both formulations. In PD rat models, the administration of both formulations positively influenced the behavioral functions of the animals. Interpretation: Our findings suggest that the administration of both CMA1 and CMA2 markedly enhanced metabolic and behavioral outcomes, aligning with neuro-histological observations. These findings underscore the promise of CMA2 administration as an effective therapeutic strategy for enhancing metabolic parameters and cognitive function in AD and PD patients.

Place, publisher, year, edition, pages
MDPI AG, 2024
Keywords
Alzheimer’s disease, animal models, combined metabolic activators, Parkinson’s disease
National Category
Neurosciences
Identifiers
urn:nbn:se:kth:diva-346370 (URN)10.3390/biomedicines12040927 (DOI)001210272400001 ()2-s2.0-85191714229 (Scopus ID)
Note

QC 20240514

Available from: 2024-05-14 Created: 2024-05-14 Last updated: 2024-05-14Bibliographically approved
Iqbal, S., Sebhaoui, J., Ashraf, S., Özcan, M., Kim, W., Belmen, B., . . . Mardinoglu, A. (2024). Design and synthesis of novel JNK inhibitors targeting liver pyruvate kinase for the treatment of non-alcoholic fatty liver disease and hepatocellular carcinoma. Bioorganic chemistry, 147, Article ID 107425.
Open this publication in new window or tab >>Design and synthesis of novel JNK inhibitors targeting liver pyruvate kinase for the treatment of non-alcoholic fatty liver disease and hepatocellular carcinoma
Show others...
2024 (English)In: Bioorganic chemistry, ISSN 0045-2068, Vol. 147, article id 107425Article in journal (Refereed) Published
Abstract [en]

Non-alcoholic fatty liver disease (NAFLD) comprises a broad range of liver disease including hepatocellular carcinoma (HCC) with is no FDA-approved drug. Liver pyruvate kinase (PKL) is a major regulator of metabolic flux and ATP generation in liver presenting a potential target for the treatment of NAFLD. Based on our recent finding of JNK-5A's effectiveness in inhibiting PKLR expression through a drug repositioning pipeline, this study aims to improve its efficacy further. We synthesized a series of JNK-5A analogues with targeted modifications, guided by molecular docking studies. These compounds were evaluated for their activities on PKL expression, cell viability, triacylglyceride (TAG) levels, and the expressions of steatosis-related proteins in the human HepG2 cell line. Subsequently, the efficacy of these compounds was assessed in reducing TAG level and toxicity. Compounds 40 (SET-151) and 41 (SET-152) proved to be the most efficient in reducing TAG levels (11.51 ± 0.90 % and 10.77 ± 0.67 %) and demonstrated lower toxicity (61.60 ± 5.00 % and 43.87 ± 1.42 %) in HepG2 cells. Additionally, all synthesized compounds were evaluated for their anti-cancer properties revealing that compound 74 (SET-171) exhibited the highest toxicity in cell viability with IC50 values of 8.82 µM and 2.97 µM in HepG2 and Huh7 cell lines, respectively. To summarize, compounds 40 (SET-151) and 41 (SET-152) show potential for treating NAFLD, while compound 74 (SET-171) holds potential for HCC therapy.

Place, publisher, year, edition, pages
Elsevier BV, 2024
Keywords
HCC, JNK-IN-5A, Liver pyruvate kinase, NAFLD, TAG level
National Category
Gastroenterology and Hepatology
Identifiers
urn:nbn:se:kth:diva-346508 (URN)10.1016/j.bioorg.2024.107425 (DOI)2-s2.0-85192210590 (Scopus ID)
Note

QC 20240520

Available from: 2024-05-16 Created: 2024-05-16 Last updated: 2025-02-11Bibliographically 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
Iqbal, S., Islam, M. Z., Ashraf, S., Kim, W., AL-Sharabi, A. A., Ozcan, M., . . . Mardinoglu, A. (2024). Discovery of Cell-Permeable Allosteric Inhibitors of Liver Pyruvate Kinase: Design and Synthesis of Sulfone-Based Urolithins. International Journal of Molecular Sciences, 25(14), Article ID 7986.
Open this publication in new window or tab >>Discovery of Cell-Permeable Allosteric Inhibitors of Liver Pyruvate Kinase: Design and Synthesis of Sulfone-Based Urolithins
Show others...
2024 (English)In: International Journal of Molecular Sciences, ISSN 1661-6596, E-ISSN 1422-0067, Vol. 25, no 14, article id 7986Article in journal (Refereed) Published
Abstract [en]

Metabolic dysfunction-associated fatty liver disease (MAFLD) presents a significant global health challenge, characterized by the accumulation of liver fat and impacting a considerable portion of the worldwide population. Despite its widespread occurrence, effective treatments for MAFLD are limited. The liver-specific isoform of pyruvate kinase (PKL) has been identified as a promising target for developing MAFLD therapies. Urolithin C, an allosteric inhibitor of PKL, has shown potential in preliminary studies. Expanding upon this groundwork, our study delved into delineating the structure-activity relationship of urolithin C via the synthesis of sulfone-based urolithin analogs. Our results highlight that incorporating a sulfone moiety leads to substantial PKL inhibition, with additional catechol moieties further enhancing this effect. Despite modest improvements in liver cell lines, there was a significant increase in inhibition observed in HepG2 cell lysates. Specifically, compounds 15d, 9d, 15e, 18a, 12d, and 15a displayed promising IC50 values ranging from 4.3 µM to 18.7 µM. Notably, compound 15e not only demonstrated a decrease in PKL activity and triacylglycerol (TAG) content but also showed efficient cellular uptake. These findings position compound 15e as a promising candidate for pharmacological MAFLD treatment, warranting further research and studies.

Place, publisher, year, edition, pages
MDPI AG, 2024
Keywords
allosteric PKL inhibition, cell permeability, MAFLD, pyruvate kinase liver, sulfone-based urolithin analogues, TAG content
National Category
Biochemistry Molecular Biology
Identifiers
urn:nbn:se:kth:diva-351706 (URN)10.3390/ijms25147986 (DOI)001277675400001 ()39063228 (PubMedID)2-s2.0-85199777848 (Scopus ID)
Note

QC 20240814

Available from: 2024-08-13 Created: 2024-08-13 Last updated: 2025-02-20Bibliographically approved
Mohammadi, E., Dashti, S., Shafizade, N., Jin, H., Zhang, C., Lam, S., . . . Sekhavati, M. H. (2024). Drug repositioning for immunotherapy in breast cancer using single-cell analysis. npj Systems Biology and Applications, 10(1), Article ID 37.
Open this publication in new window or tab >>Drug repositioning for immunotherapy in breast cancer using single-cell analysis
Show others...
2024 (English)In: npj Systems Biology and Applications, E-ISSN 2056-7189, Vol. 10, no 1, article id 37Article in journal (Refereed) Published
Abstract [en]

Immunomodulatory peptides, while exhibiting potential antimicrobial, antifungal, and/or antiviral properties, can play a role in stimulating or suppressing the immune system, especially in pathological conditions like breast cancer (BC). Thus, deregulation of these peptides may serve as an immunotherapeutic strategy to enhance the immune response. In this meta-analysis, we utilized single-cell RNA sequencing data and known therapeutic peptides to investigate the deregulation of these peptides in malignant versus normal human breast epithelial cells. We corroborated our findings at the chromatin level using ATAC-seq. Additionally, we assessed the protein levels in various BC cell lines. Moreover, our in-house drug repositioning approach was employed to identify potential drugs that could positively impact the relapse-free survival of BC patients. Considering significantly deregulated therapeutic peptides and their role in BC pathology, our approach aims to downregulate B2M and SLPI, while upregulating PIGR, DEFB1, LTF, CLU, S100A7, and SCGB2A1 in BC epithelial cells through our drug repositioning pipeline. Leveraging the LINCS L1000 database, we propose BRD-A06641369 for B2M downregulation and ST-4070043 and BRD-K97926541 for SLPI downregulation without negatively affecting the MHC complex as a significantly correlated pathway with these two genes. Furthermore, we have compiled a comprehensive list of drugs for the upregulation of other selected immunomodulatory peptides. Employing an immunotherapeutic approach by integrating our drug repositioning pipeline with single-cell analysis, we proposed potential drugs and drug targets to fortify the immune system against BC.

Place, publisher, year, edition, pages
Springer Nature, 2024
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:kth:diva-345962 (URN)10.1038/s41540-024-00359-z (DOI)001198637300001 ()38589404 (PubMedID)2-s2.0-85190320576 (Scopus ID)
Note

QC 20240430

Available from: 2024-04-30 Created: 2024-04-30 Last updated: 2024-08-30Bibliographically approved
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0002-3721-8586

Search in DiVA

Show all publications