kth.sePublications
Change search
Link to record
Permanent link

Direct link
Kim, Woonghee
Publications (10 of 32) Show all publications
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
El Hafi, M., Bouzian, Y., Parvizi, N., Kim, W., Subaşioğlu, M., Ozcan, M., . . . Mardinoglu, A. (2025). Synthesis and biological assessment of BUB1B inhibitors for the treatment of clear cell renal cell carcinoma. European Journal of Medicinal Chemistry, 285, Article ID 117247.
Open this publication in new window or tab >>Synthesis and biological assessment of BUB1B inhibitors for the treatment of clear cell renal cell carcinoma
Show others...
2025 (English)In: European Journal of Medicinal Chemistry, ISSN 0223-5234, E-ISSN 1768-3254, Vol. 285, article id 117247Article in journal (Refereed) Published
Abstract [en]

Clear cell renal cell carcinoma (ccRCC) presents substantial therapeutic challenges due to its molecular heterogeneity, limited response to conventional therapies, and widespread drug resistance. Recent advancements in molecular research have identified novel targets, such as BUB1B, which has been identified through global transcriptomic profiling and gene co-expression network analysis as critical in ccRCC progression. In this study, we synthesized 40 novel derivatives of TG-101209 to modulate BUB1B expression and activity, leading to the induction of apoptosis in Caki-1 cells. The molecular structures of all compounds were confirmed via 1H and 13C NMR and mass spectrometry. Computational docking studies were conducted using Schrödinger Maestro software. The efficacy of the compounds on cell viability was screened using the MTT assay and further validated by the LDH assay. The expression of the target protein BUB1B and apoptosis-related proteins was analyzed via western blotting. BUB1B activity was assessed through an enzymatic assay, and compound binding efficacy was evaluated using a cellular thermal shift assay (CETSA). The results indicated that four compounds (7h, 8h, 8i, and 8j) demonstrate stronger molecular interactions and better conformational fit within the target cavity, leading to improved binding affinity. These compounds also exhibited more potency in reducing the viability of Caki-1 cells compared to TG-101209. In particular, compound 8h was identified as the most effective, exhibiting the strongest inhibitory effect on BUB1B and inducing apoptosis. Compound 8h demonstrated intracellular binding with BUB1B, similar to TG-101209, but through a different binding moiety that destabilizes the BUB1B protein structure, whereas TG-101209 stabilizes it. In conclusion, compound 8h, by destabilizing BUB1B and inducing apoptosis, shows promise as a potent therapeutic candidate for clear cell renal cell carcinoma (ccRCC) treatment.

Place, publisher, year, edition, pages
Elsevier BV, 2025
Keywords
BUB1B, Caki-1, ccRCC, Drug design, Structure-activity relationship, TG-101209
National Category
Cancer and Oncology Pharmaceutical Sciences Pharmacology and Toxicology
Identifiers
urn:nbn:se:kth:diva-358891 (URN)10.1016/j.ejmech.2025.117247 (DOI)001423233400001 ()39818011 (PubMedID)2-s2.0-85214836991 (Scopus ID)
Note

QC 20250303

Available from: 2025-01-23 Created: 2025-01-23 Last updated: 2025-03-03Bibliographically approved
Bouzian, Y., El Hafi, M., Parvizi, N., Kim, W., Subaşioğlu, M., Ozcan, M., . . . Mardinoglu, A. (2024). Design and evaluation of novel inhibitors for the treatment of clear cell renal cell carcinoma. Bioorganic chemistry, 151, Article ID 107597.
Open this publication in new window or tab >>Design and evaluation of novel inhibitors for the treatment of clear cell renal cell carcinoma
Show others...
2024 (English)In: Bioorganic chemistry, ISSN 0045-2068, Vol. 151, article id 107597Article in journal (Refereed) Published
Abstract [en]

The efficacy of conventional chemotherapies in treating clear cell renal cell carcinoma (ccRCC) is often limited due to its high molecular diversity, generally low response rates to standard treatments, and prevalent drug resistance. Recent advancements in the molecular understanding of ccRCC, alongside the discovery of novel therapeutic agents targeting specific proteins, have significantly altered the treatment landscape for ccRCC. Here, we synthesized 27 new compounds that are derivatives of TG-101209 to modulate BUB1B (BUB1 mitotic checkpoint serine/threonine kinase B). BUB1B has been recently identified as a drug target for the development of effective ccRCC treatment based on global transcriptomics profiling of ccRCC tumours and gene co-expression network analysis. We characterized the molecular structures of these 27 compounds by 1H and 13C NMR and Mass spectrometry. We evaluated the effect of these 27 compounds by analysing the modulation of the BUB1B expression. Our primary objective was to design and assess the efficacy of these new compounds in reducing the viability of Caki-1 cells, a ccRCC cell line. We performed the computational docking studies by the Schrödinger Maestro software and demonstrated that three of these compounds (13a, 5i, and 5j) effectively downregulated BUB1B expression and eventually triggered necrosis and apoptosis in the Caki-1 cell line based on the structure–activity relationship (SAR) analysis. The IC50 values for compounds 13a, 5i, and 5j were calculated as 2.047 µM, 10.046 µM, and 6.985 µM, respectively, indicating their potent inhibitory effects on cell viability. Our study suggests that these compounds targeting BUB1B could offer a more effective and promising approach for ccRCC treatment compared to the conventionally used tyrosine kinase inhibitors. Our study underscores the potential of leveraging targeted therapies against specific molecular pathways in ccRCC may open new avenues for the development of effective treatment strategies against ccRCC.

Place, publisher, year, edition, pages
Elsevier BV, 2024
Keywords
BUB1B, Caki-1, ccRCC, Drug design, TG-101209
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:kth:diva-350979 (URN)10.1016/j.bioorg.2024.107597 (DOI)001271032500001 ()39002511 (PubMedID)2-s2.0-85198275564 (Scopus ID)
Note

QC 20240724

Available from: 2024-07-24 Created: 2024-07-24 Last updated: 2025-05-27Bibliographically 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)38714117 (PubMedID)2-s2.0-85192210590 (Scopus ID)
Note

QC 20240520

Available from: 2024-05-16 Created: 2024-05-16 Last updated: 2025-05-27Bibliographically 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
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
Yang, H., Zhang, C., Kim, W., Shi, M., Kiliclioglu, M., Bayram, C., . . . Mardinoglu, A. (2024). Multi-tissue network analysis reveals the effect of JNK inhibition on dietary sucrose-induced metabolic dysfunction in rats.
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...
2024 (English)Manuscript (preprint) (Other academic)
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 modelling 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.

National Category
Basic Medicine
Identifiers
urn:nbn:se:kth:diva-346361 (URN)10.1101/2024.04.22.590583 (DOI)
Note

QC 20240514

Available from: 2024-05-13 Created: 2024-05-13 Last updated: 2024-06-13Bibliographically 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
Matić, J., Akladios, F., Battisti, U. M., Håversen, L., Nain-Perez, A., Füchtbauer, A. F., . . . Grøtli, M. (2024). Sulfone-based human liver pyruvate kinase inhibitors – Design, synthesis and in vitro bioactivity. European Journal of Medicinal Chemistry, 269, Article ID 116306.
Open this publication in new window or tab >>Sulfone-based human liver pyruvate kinase inhibitors – Design, synthesis and in vitro bioactivity
Show others...
2024 (English)In: European Journal of Medicinal Chemistry, ISSN 0223-5234, E-ISSN 1768-3254, Vol. 269, article id 116306Article in journal (Refereed) Published
Abstract [en]

Non-alcoholic fatty liver disease (NAFLD) is a prevalent pathological condition characterised by the accumulation of fat in the liver. Almost one-third of the global population is affected by NAFLD, making it a significant health concern. However, despite its prevalence, there is currently no approved drug specifically designed for the treatment of NAFLD. To address this critical gap, researchers have been investigating potential targets for NAFLD drug development. One promising candidate is the liver isoform of pyruvate kinase (PKL). In recent studies, Urolithin C, an allosteric inhibitor of PKL, has emerged as a potential lead compound for therapeutic intervention. Building upon this knowledge, our team has conducted a comprehensive structure-activity relationship of Urolithin C. In this work, we have employed a scaffold-hopping approach, modifying the urolithin structure by replacing the urolithin carbonyl with a sulfone moiety. Our structure-activity relationship analysis has identified the sulfone group as particularly favourable for potent PKL inhibition. Additionally, we have found that the presence of catechol moieties on the two aromatic rings further improves the inhibitory activity. The most promising inhibitor from this new series displayed nanomolar inhibition, boasting an IC50 value of 0.07 μM. This level of potency rivals that of urolithin D and significantly surpasses the effectiveness of urolithin C by an order of magnitude. To better understand the molecular interactions underlying this inhibition, we obtained the crystal structure of one of the inhibitors complexed with PKL. This structural insight served as a valuable reference point, aiding us in the design of inhibitors.

Place, publisher, year, edition, pages
Elsevier BV, 2024
Keywords
Allosteric PKL modulation, NAFLD, PKL inhibitors, Pyruvate kinase liver, Sulfone heterocycles
National Category
Biochemistry Molecular Biology
Identifiers
urn:nbn:se:kth:diva-344582 (URN)10.1016/j.ejmech.2024.116306 (DOI)001209771400001 ()38471358 (PubMedID)2-s2.0-85187492456 (Scopus ID)
Note

QC 20240513

Available from: 2024-03-20 Created: 2024-03-20 Last updated: 2025-02-20Bibliographically approved
Ashraf, S., Iqbal, S., Sebhaoui, J., Özcan, M., Kim, W., Belmen, B., . . . Mardinoglu, A. (2024). Synthesis, spectroscopic characterization, DFT and molecular docking of N-(3-cyano-4,5,6,7-tetrahydrobenzo[b]thiophen-2-yl) naphthalene-1-sulfonamide derivatives. Journal of Molecular Structure, 1312, Article ID 138470.
Open this publication in new window or tab >>Synthesis, spectroscopic characterization, DFT and molecular docking of N-(3-cyano-4,5,6,7-tetrahydrobenzo[b]thiophen-2-yl) naphthalene-1-sulfonamide derivatives
Show others...
2024 (English)In: Journal of Molecular Structure, ISSN 0022-2860, E-ISSN 1872-8014, Vol. 1312, article id 138470Article in journal (Refereed) Published
Abstract [en]

Liver pyruvate kinase (PKL) is a key player in controlling metabolic pathways and ATP production within the liver's glycolysis pathway. Since PKL modulators have been identified as a promising target for treating hepatocellular carcinoma (HCC) and non-alcoholic fatty liver disease (NAFLD), our research is centered on the development and synthesis of derivatives of N-(3-cyano-4,5,6,7-tetrahydrobenzo[b]thiophen-2-yl) naphthalene-1-sulfonamide with the aim of modulating PLK. To improve PKL specificity, we used structural analysis and modeling as a guide. Notably, compound PKL-05 became the series' only active ingredient. DFT, Hirshfeld surface analysis, and molecular docking were used in our study to thoroughly examine the connection between compound structures and their computational functions. The global hardness and softness energy values, as well as the HOMO-LUMO energy gap value, were computed in order to forecast the chemical reactivity of this newly synthesized molecule. These energy values indicate that this molecule tends to be chemically stable and has little chemical reactivity. The results demonstrated a strong agreement between theoretical forecasts and experimental findings. In particular, PKL-05 exhibits encouraging traits that establish it as a useful starting point for additional research in the search for innovative PKL modulators to tackle the treatment issues associated with NAFLD and HCC.

Place, publisher, year, edition, pages
Elsevier B.V., 2024
Keywords
DFT, Docking, Hirschfield surface analysis, Liver pyruvate kinase, Synthesis, X-ray
National Category
Biochemistry Molecular Biology
Identifiers
urn:nbn:se:kth:diva-346494 (URN)10.1016/j.molstruc.2024.138470 (DOI)001240351100001 ()2-s2.0-85192494249 (Scopus ID)
Note

QC 20240620

Available from: 2024-05-16 Created: 2024-05-16 Last updated: 2025-02-20Bibliographically approved
Organisations

Search in DiVA

Show all publications