1
|
Fuenteslópez CV, Papapavlou M, Thompson MS, Ye H. Engineering a long-lasting microvasculature in vitro model for traumatic injury research. BIOMATERIALS ADVANCES 2025; 174:214310. [PMID: 40220460 DOI: 10.1016/j.bioadv.2025.214310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 02/21/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
Microvascular injuries can have systemic physiological effects that exacerbate other injuries and pose a danger to life. Reliable in vitro microvascular models are required to enhance understanding of traumatic injuries. This research aims to develop and optimise a three-dimensional (3D) hydrogel construct for the formation and long-term stability of an in vitro microvascular model for trauma research. First, we develop a 3D hydrogel scaffold using a physiologically relevant cell type to enable the formation of a durable microvascular endothelial network and validate it against the gold standard: HUVECs. Then, we explore the impact of modifying the hydrogel composition, specifically fibrinogen source and concentration, medium, and crosslinking ratio, on scaffold material properties and, consequently, the formation of endothelial networks, their architecture, and long-term integrity. Our results demonstrate that 3D hydrogel scaffolds are crucial for maintaining network stability beyond the initial 24 h. For trauma research applications, the material properties and mechanical behaviour of the hydrogels are critical. Microrheometry revealed that fibrinogen concentration significantly influences gelation times, absorbance rate, storage modulus (G'), loss modulus (G"), and complex viscosity, while also reducing creep compliance. Our multi-pronged approach to engineering microvasculature constructs revealed that variations in hydrogel composition, including fibrinogen concentration and source, crosslinking ratio and choice of medium, strongly affect the hydrogel material characteristics and, in turn, the resulting microvascular networks. Hydrogels made with high concentrations of human fibrinogen, a 200:10:1 crosslinking ratio, and endothelial basal medium (EBM) or EBM supplemented with VEGF performed best, demonstrating superior long-term network stability. The microvasculature construct developed here could be used as a potential platform for studying traumatic injuries, as well as testing interventions aimed at improving recovery and mitigating damage.
Collapse
Affiliation(s)
- Carla Verónica Fuenteslópez
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| | - Mariella Papapavlou
- Department of Materials, University of Oxford, Parks Road, Oxford OX1 3PH, United Kingdom
| | - Mark S Thompson
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| |
Collapse
|
2
|
Panja S, Kapoor E, Siddhanta K, Jogdeo CM, Sil D, Khan RI, Kumari N, Ding L, Gendelman HE, Singh AB, Oupický D. Bioactive polymers as stimulus-responsive anti-metastatic combination agents to treat pancreatic cancer. Biomaterials 2025; 320:123255. [PMID: 40107179 DOI: 10.1016/j.biomaterials.2025.123255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
The intractable and devastating nature of pancreatic ductal adenocarcinoma (PDAC) necessitates an urgent need for novel therapies. This study presents the development of a novel polymer prodrug system for the combination treatment of PDAC, based on an optimized pharmacologically active anti-metastatic macromolecular carrier, PCQ, conjugated with gemcitabine (GEM). Structure-activity relationship evaluations showed that random PCQ copolymers exhibited superior anti-migratory activity compared to the gradient PCQ analogs. GEM was incorporated into the random PCQ copolymers using disulfide linker to prepare a reduction-responsive prodrug, PCQ(r)6-SS-GEM12. The resultant therapeutic system presents a pharmacologically active delivery strategy that targets both the proliferative and the metastatic phenotype in PDAC. The PCQ(r)6-SS-GEM12 prodrug demonstrated a selective release of GEM under the reductive tumor environment leading to a significant inhibition of tumor growth with pronounced anti-metastatic effect. Collectively, our data show that the combination of anti-metastatic PCQ and cytotoxic GEM-based reduction-responsive prodrug polymer offers an innovative strategy to treat PDAC.
Collapse
Affiliation(s)
- Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA; Department of Pharmacology and Experimental Neuroscience, USA
| | - Ekta Kapoor
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Diptesh Sil
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Rubayat I Khan
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Neha Kumari
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | | | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA.
| |
Collapse
|
3
|
Prasad KN, C C, Karthik Y, Girish GV, A S. The future of lactoferrin: A closer look at LipoDuo technology. J Liposome Res 2025; 35:153-158. [PMID: 39799475 DOI: 10.1080/08982104.2025.2451235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/23/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Lactoferrin (Lf), a multifunctional glycoprotein known for its roles in immune modulation, iron metabolism, and antimicrobial activity, has limited therapeutic efficacy due to poor bioavailability. Liposomal encapsulation of lactoferrin (LLf) offers a potential solution by improving its stability, absorption, and sustained release, making it a promising candidate for various clinical applications. This study aims to compare the effectiveness of LLf and plain Lf in cellular uptake, proliferation, and wound healing using HEK-293T and Caco-2 cell lines. METHODS Cell uptake, proliferation, and wound healing assays were conducted using HEK-293T and Caco-2 cells to evaluate the bioavailability and therapeutic efficacy of LLf compared to plain Lf. The cellular uptake was assessed over a 24-h period using an indirect ELISA method. Cell proliferation was measured using the MTT assay, while wound healing was evaluated using a scratch assay to observe cell migration over 48 h. RESULTS LLf demonstrated significantly higher cellular uptake in both HEK-293T and Caco-2 cells, with peak internalization at 4 h, compared to plain Lf. In proliferation studies, LLf showed a dose-dependent increase in cell growth, achieving a 71% proliferation rate at 75 µg/mL, while plain Lf reached only 53%. LLf also accelerated wound healing, with nearly complete closure by 48 h, compared to 51.3% closure with plain Lf. CONCLUSION The results indicate that liposomal encapsulation significantly enhances lactoferrin's bioavailability, proliferation-inducing capacity, and wound healing efficacy. LLf's superior performance in these key areas suggests its potential for broader therapeutic applications, particularly in wound care, immune modulation, and tissue regeneration. Future clinical studies are warranted to validate the therapeutic benefits of LLf in vivo.
Collapse
Affiliation(s)
- K N Prasad
- Samarth Biorigins LLP, KIADB Industrial Area, Tumkur, India
| | - Chaithra C
- Samarth Biorigins LLP, KIADB Industrial Area, Tumkur, India
| | - Yalpi Karthik
- Samarth Biorigins LLP, KIADB Industrial Area, Tumkur, India
| | - G V Girish
- Samarth Biorigins LLP, KIADB Industrial Area, Tumkur, India
| | - Sandhya A
- Samarth Biorigins LLP, KIADB Industrial Area, Tumkur, India
| |
Collapse
|
4
|
Maloba GO, Were T, Barasa E, Mohamed N, Arshi A, Gallyas F. Synergistic Effects of 2-Deoxyglucose and Diclofenac Sodium on Breast Cancer Cells: A Comparative Evaluation of MDA-231 and MCF7 Cells. Int J Mol Sci 2025; 26:4894. [PMID: 40430033 PMCID: PMC12112485 DOI: 10.3390/ijms26104894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Resistance of breast cancers to chemotherapy remains a global challenge to date. Drug combination studies between anti-cancer agents are increasingly becoming therapeutic strategies, geared towards alleviating breast cancers. Previously, 2-deoxyglucose has been shown to target and interrupt glycolysis. Available evidence also suggests that diclofenac, which was originally designed as a pain reliever, could inhibit the proliferation of breast cancer cells. However, the reverse Warburg effect and other metabolic reprogramming mechanisms in breast cancers limit the pharmacological application of both 2-deoxyglucose and diclofenac as mono-therapeutic agents. The present study explores the additive anti-cancer effects of 2-deoxyglucose and diclofenac sodium on breast cancer cells. In this study, MDA-231 and MCF7 cells were treated with 2-deoxyglucose and diclofenac sodium in single and combination doses before being evaluated for viability, cell growth, reactive oxygen species, apoptotic and necrotic phases, and migration abilities. Additionally, immunoblotting of pro-apoptotic proteins, Caspase-3 and Caspase-9, and a hypoxia-inducible factor-1 alpha, was also performed. The results showed that combination treatments of the cells with the drugs exhibited additive anti-cancer effects by limiting proliferation, enhancing cytotoxic reactive oxygen species generation, enhancing apoptosis and necrosis, limiting colony formation and expansion of cells, and inhibiting cell migration. The degrees of cytotoxicity of combined treatments were almost similar in both cell lines, although with minimal differences. Put together, these results reveal the novel synergistic effects of 2-deoxyglucose and diclofenac sodium on breast cancer cells, hence potentially elevating their pharmacological profile in the overall breast cancer therapy.
Collapse
Affiliation(s)
- Geofrey Ouma Maloba
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary; (G.O.M.); (N.M.); (A.A.)
| | - Tom Were
- Department of Pathology, Masinde Muliro University of Science and Technology, Kakamega 190-50100, Kenya; (T.W.); (E.B.)
| | - Erick Barasa
- Department of Pathology, Masinde Muliro University of Science and Technology, Kakamega 190-50100, Kenya; (T.W.); (E.B.)
| | - Nasreldeen Mohamed
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary; (G.O.M.); (N.M.); (A.A.)
| | - Arshi Arshi
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary; (G.O.M.); (N.M.); (A.A.)
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary; (G.O.M.); (N.M.); (A.A.)
| |
Collapse
|
5
|
Ribeiro JRL, Calheiros J, Silva RAM, Gonçalves BMF, Afonso CAM, Saraiva L, Ferreira MJU. Exploring the Anticancer Properties of 1,2,3-Triazole-Substituted Andrographolide Derivatives. Pharmaceuticals (Basel) 2025; 18:750. [PMID: 40430567 PMCID: PMC12114840 DOI: 10.3390/ph18050750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: The search for new anticancer agents from natural sources remains a key strategy in drug discovery. This study aimed to synthesize and evaluate novel triazole derivatives of the diterpenic lactone andrographolide for their antiproliferative activity against various cancer cell lines. Methods: Twenty-two new triazole derivatives (5-26), of the triacetyl derivative (2) of the diterpenic lactone andrographolide (1), were synthesized via the azide-alkyne "click reaction". The antiproliferative effects of compounds 1-26 were evaluated using the sulforhodamine B assay against a panel of cancer cell lines and a non-tumorigenic colon cell line. A representative compound, triazole derivative 12, was further evaluated in human pancreatic ductal adenocarcinoma (PANC-1) cells for its effects on the cell cycle, apoptosis, migration, and drug synergy with 5-fluorouracil. Results: Several compounds, specifically, 9, 14, 16, and 17, bearing a phenyl moiety, exhibited improved antiproliferative activity compared to the parental compound 1. Derivative 12, selected for further investigation, induced G2/M cell cycle arrest and apoptosis in a concentration-dependent manner. Additionally, this compound significantly reduced cell migration and demonstrated synergistic effects with 5-fluorouracil in PANC-1 cells. Conclusions: The synthesized andrographolide-based triazole derivatives, particularly compound 12, showed promising antiproliferative activity and mechanisms relevant to cancer therapy. These findings support their potential as lead compounds for further development in anticancer research.
Collapse
Affiliation(s)
- Joana R. L. Ribeiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Juliana Calheiros
- LAQV/REQUIMTE-Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, Universidade do Porto, 4050-313 Porto, Portugal
| | - Rita A. M. Silva
- LAQV/REQUIMTE-Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, Universidade do Porto, 4050-313 Porto, Portugal
| | - Bruno M. F. Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Carlos A. M. Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Lucília Saraiva
- LAQV/REQUIMTE-Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, Universidade do Porto, 4050-313 Porto, Portugal
| | - Maria-José U. Ferreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
6
|
Vengadesan V, Muniyandi J, Yadav N, G P, Sawant AR, Sankaranarayanan L, K P, Swu T, Sadras SR. Borrelidin M: a new borrelidin derivative obtained from Streptomyces rochei VL-16 exhibited potent antibacterial and antiproliferative properties. Int Microbiol 2025:10.1007/s10123-025-00673-4. [PMID: 40372540 DOI: 10.1007/s10123-025-00673-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/30/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
The lookout for novel metabolites to overcome the threat imposed by antibiotic resistant pathogens is on the rise globally. The present study reports on the identification and characterisation of a new borrelidin derivative from Streptomyces rochei VL-16 strain that was isolated from the soil samples of Osudu Lake in Puducherry. The active compound produced by Streptomyces rochei VL-16 strain was purified sequentially by thin layer, column and high performance liquid chromatography techniques. The purified compound was subsequently identified as borrelidin through Q-TOF-HR-LCMS, FTIR and UV-Vis spectroscopy techniques. Based on integration proton NMR signals and HR-ESI-MS peak pattern, the compound was further identified as a new derivative of borrelidin. Its 3D structure was optimised using density functional theory. The new derivative obtained in this study was designated as borrelidin M (C28H41NO5, Mol. wt.471.3 g/mole). This derivative was found to exhibit potent inhibitory effects against the foodborne pathogens including Clostridium perfringens, Aeromonas hydrophila, Staphylococcus aureus, Listeria monocytogenes and Yersinia enterocolitica. Also, borrelidin M exhibited anticancer and anti-metastatic effects against A549 cell line with an IC50 value of 17.5 μM. Collectively, these findings indicated promising biomedical applications of this new derivative of borrelidin.
Collapse
Affiliation(s)
- Vinoth Vengadesan
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Jagan Muniyandi
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Neha Yadav
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Premkumar G
- Department of Chemistry, Pondicherry University, Puducherry, India
| | | | | | - Prashanth K
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Toka Swu
- Department of Chemistry, Pondicherry University, Puducherry, India
| | - Sudha Rani Sadras
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India.
| |
Collapse
|
7
|
Margariti M, Roussaki M, Sakellis E, Boukos N, Theodosiou M, Kostopoulou N, Efthimiadou EK. Dextran-Assisted Biomimetic Fabrication of CdSe:Eu QDs With Enhanced Biocompatibility and Stability. Chemistry 2025:e202500986. [PMID: 40328676 DOI: 10.1002/chem.202500986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/22/2025] [Accepted: 05/06/2025] [Indexed: 05/08/2025]
Abstract
A dextran templated biomimetic synthetic approach was applied for the preparation of Cadmium Selinium:Europium Quantum Dots (CdSe:Eu QDs). The present work examines synthetic variables including the effect of increasing Europium concentration during QDs' synthesis and the use of different coating agents (citrate versus APTES) to improve the QDs' colloidal stability and ameliorate their cytotoxic behavior. All samples were subjected to structural and morphological characterizations via Fourier Transform infrared (FT-IR) spectroscopy, UV-Visible (UV-Vis) spectrophotometry, Transmission Electron Microscopy (TEM), and Dynamic Light Scattering (DLS). Subsequently, biological evaluation of all the synthesized samples of bare and APTES coated CdSe:Eu QDs was conducted on epithelial HaCaT and breast cancer MCF-7. Wound healing assays were conducted to assess cytotoxicity and cell migration under QD treatment. Results showed that both coated and uncoated QDs influenced wound closure rates in a concentration- and time-dependent manner. Finally, fluorescence microscopy revealed significant green luminescence of CdSe:Eu quantum dots (QDs) in both HaCaT and MCF-7 cell lines, indicating efficient cellular uptake. The internalization of QDs was influenced by their surface properties and charge, suggesting an endocytic uptake pathway.
Collapse
Affiliation(s)
- Myrto Margariti
- Inorganic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15771, Greece
| | - Marianna Roussaki
- Inorganic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15771, Greece
| | - Elias Sakellis
- Institute of Nanoscience and Nanotechnology, National Center of Scientific Research "Demokritos", Agia Paraskevi, 15341, Greece
- Section of Condensed Matter Physics, Department of Physics, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - Nikos Boukos
- Institute of Nanoscience and Nanotechnology, National Center of Scientific Research "Demokritos", Agia Paraskevi, 15341, Greece
| | - Maria Theodosiou
- Inorganic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15771, Greece
| | - Nikoleta Kostopoulou
- Inorganic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15771, Greece
| | - Eleni K Efthimiadou
- Inorganic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15771, Greece
| |
Collapse
|
8
|
Hao X, Chen Y, Sahu D, Przanowska RK, Aaiyas M, Weidmann CA, Nardi I, Weeks KM, Dutta A. A 36-base hairpin within lncRNA DRAIC, which is modulated by alternative splicing, interacts with the IKKα coiled-coil domain and inhibits NF-κB and tumor cell phenotypes. J Biol Chem 2025:110172. [PMID: 40320073 DOI: 10.1016/j.jbc.2025.110172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/08/2025] Open
Abstract
A tumor-suppressive long noncoding RNA (lncRNA) DRAIC (down-regulated RNA in cancers) inhibits NF-κB activity and physically interacts with IKKα, a kinase component of the IKK complex, in several cancer types. Here we explore the precise molecular mechanism involved in this interaction and suppression. Using SHAPE-MaP, we identified a 36-nucleotide hairpin (A+B) within DRAIC that is necessary and sufficient for its anti-oncogenic function. RNA immunoprecipitation (RIP) and Electrophoretic mobility shift assays (EMSA) confirmed this hairpin physically interacts with the coiled coil domain of IKKα. A+B RNA has a high binding affinity (KD ∼1-7 nM) to the coiled-coil domain of IKKα. The binding of A+B disrupts the dimerization of NEMO and IKKα coiled-coil domains, a critical step for IKK action. Consistent with this, A+B inhibits the phosphorylation of the NF-κB inhibitor IκBα and suppresses NF-κB activity. Publicly available tumor RNAseq data revealed that alternative splicing modulates the presence of this critical hairpin: the inclusion of exon 4a (encoding one side of the A+B hairpin) in lung tumors correlates with reduced NF-κB activity. By demonstrating that the A+B hairpin is both necessary and sufficient to inhibit IKK and oncogenic phenotypes, this study underscores the centrality of IKKα interaction and NF-κB inhibition in DRAIC-mediated cancer suppression and indicates that the activity of this lncRNA is regulated by alternative splicing. This study also reveals the first example of a short RNA disrupting coiled-coil dimerization, offering a new approach to disrupt such dimerization in cancer biology.
Collapse
Affiliation(s)
- Xiaoxiao Hao
- University of Alabama at Birmingham, Department of Genetics, Birmingham, AL
| | - Yuechuan Chen
- University of Alabama at Birmingham, Department of Genetics, Birmingham, AL
| | - Divya Sahu
- University of Alabama at Birmingham, Department of Genetics, Birmingham, AL
| | - Róża K Przanowska
- University of Virginia, Department of Biochemistry and Molecular Genetics, Charlottesville, VA
| | - Mujawar Aaiyas
- University of Alabama at Birmingham, Department of Genetics, Birmingham, AL
| | - Chase A Weidmann
- Department of Biological Chemistry, Center for RNA Biomedicine, Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI
| | | | - Kevin M Weeks
- Department of Chemistry, University of North Carolina, Chapel Hill, NC
| | - Anindya Dutta
- University of Alabama at Birmingham, Department of Genetics, Birmingham, AL
| |
Collapse
|
9
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation during Wound Closure. ACS Biomater Sci Eng 2025; 11:2249-2262. [PMID: 40029610 DOI: 10.1021/acsbiomaterials.4c02019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. Extra domain A (EDA) Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, EDA Fn organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional coactivator, yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate whether YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity, but on stiffer substrates, they decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest that there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
10
|
de Oliveira GGFV, Longue MF, Pescinelli LMR, Charret TS, Nogueira TSR, Pereira MTM, Vieira IJC, Abreu LS, Pascoal VDB, Pascoal ACRF. Eugenia brasiliensis: Analysis of the Chemical Profile and Evaluation of Cytotoxic Potential. Chem Biodivers 2025:e202500429. [PMID: 40219922 DOI: 10.1002/cbdv.202500429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/14/2025]
Abstract
This work evaluated the antiproliferative potential of Eugenia brasiliensis leaf extracts against the HeLa cervical cancer cell line. The extracts were prepared by maceration using hexane (EBH), dichloromethane (EBD), and ethyl acetate (EBAE), and they were evaluated for their antiproliferative potential through a 3-4,5-dimethyl-thiazol-2-yl-2,5-diphenyltetrazoliumbromide (MTT) assay in the cervical cancer cell culture (HeLa cell line) and a non-cancer cell line (NIH-3T3). EBH, EBD, and EBAE were cytotoxic in HeLa cells, with 50% inhibition concentration (IC50) = 97.59, 31.03, and 57.67 µg/mL, respectively. EBD inhibited migration and altered the cell cycle. Eight compounds were tentatively assigned to E. brasiliensis leaf extracts by interpreting their fragmentation patterns and molecular formulae obtained from mass spectra. The dichloromethane extract of the leaves of E. brasiliensis against the cells of cervical cancer showed potential cytotoxicity activity.
Collapse
Affiliation(s)
- Giovana G F V de Oliveira
- Research Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo Health Institute, Fluminense Federal University-UFF, Nova Friburgo, Brazil
| | - Milena F Longue
- Research Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo Health Institute, Fluminense Federal University-UFF, Nova Friburgo, Brazil
| | - Letícia M R Pescinelli
- Research Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo Health Institute, Fluminense Federal University-UFF, Nova Friburgo, Brazil
| | - Thiago S Charret
- Research Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo Health Institute, Fluminense Federal University-UFF, Nova Friburgo, Brazil
| | - Thalya S R Nogueira
- Laboratory of Chemical Sciences, Center for Exact Sciences and Technology, State University of North Fluminense Darcy Ribeiro-UENF, Campos dos Goytacazes, Brazil
| | - Mariana T M Pereira
- Research Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo Health Institute, Fluminense Federal University-UFF, Nova Friburgo, Brazil
| | - Ivo J C Vieira
- Laboratory of Chemical Sciences, Center for Exact Sciences and Technology, State University of North Fluminense Darcy Ribeiro-UENF, Campos dos Goytacazes, Brazil
| | - Lucas S Abreu
- Natural Products Chemistry Laboratory, Institute of Chemistry, Fluminense Federal University- UFF, Niteroi, Brazil
| | - Vinicius D B Pascoal
- Research Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo Health Institute, Fluminense Federal University-UFF, Nova Friburgo, Brazil
| | - Aislan C R F Pascoal
- Research Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo Health Institute, Fluminense Federal University-UFF, Nova Friburgo, Brazil
| |
Collapse
|
11
|
Mehanna LE, Boyd JD, Walker CG, Osborne AR, Grady ME, Berron BJ. Functional assessment of migration and adhesion to quantify cancer cell aggression. SOFT MATTER 2025; 21:2946-2957. [PMID: 40151848 DOI: 10.1039/d4sm01351d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
During epithelial-to-mesenchymal transition (EMT), cancer cells lose their cell-cell adhesion junctions as they become more metastatic, altering cell motility and focal adhesion disassembly associated with increased detachment from the primary tumor and a migratory response into nearby tissue and vasculature. Current in vitro strategies characterizing a cell's metastatic potential heavily favor quantifying the presence of cell adhesion biomarkers through biochemical analysis; however, mechanical cues such as adhesion and motility directly relate to cell metastatic potential without needing to first identify a cell specific biomarker for a particular type of cancer. This paper presents a comprehensive comparison of two functional metrics of cancer aggression, wound closure migration velocity and cell detachment from a culture surface, for three pairs of epithelial cancer cell lines (breast, endometrium, tongue tissue origins). It was found that one functional metric alone was not sufficient to categorize the cancer cell lines; instead, both metrics were necessary to identify functional trends and accurately place cells on the spectrum of metastasis. On average, cell lines with low metastatic potential (MCF-7, Ishikawa, and Cal-27) were more aggressive through wound closure migration compared to loss of cell adhesion. On the other hand, cell lines with high metastatic potential (MDA-MB-231, KLE, and SCC-25) were on average more aggressive through loss of cell adhesion compared to wound closure migration. This trend was true independent of the tissue type where the cells originated, indicating that there is a relationship between metastatic potential and the predominate type of cancer aggression. Our work presents one of the first combined studies relating cell metastatic potential to functional migration and adhesion metrics across cancer cell lines from selected tissue origins, without needing to identify tissue-specific biomarkers to achieve success. Using functional metrics provides powerful clinical relevancy for future predictive tools of cancer metastasis.
Collapse
Affiliation(s)
- Lauren E Mehanna
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY 40506, USA.
| | - James D Boyd
- Department of Mechanical and Aerospace Engineering, University of Kentucky, 151 Ralph G. Anderson Building, Lexington, KY 40506, USA.
| | - Chloe G Walker
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY 40506, USA.
| | - Adrianna R Osborne
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY 40506, USA.
| | - Martha E Grady
- Department of Mechanical and Aerospace Engineering, University of Kentucky, 151 Ralph G. Anderson Building, Lexington, KY 40506, USA.
| | - Brad J Berron
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY 40506, USA.
| |
Collapse
|
12
|
Setia M, Suvas PK, Rana M, Chakraborty A, Suvas S. Differential homing of monocytes and neutrophils in the epithelial layer of HSV-1 infected cornea regulates viral dissemination and wound healing. Ocul Surf 2025; 36:69-82. [PMID: 39793926 PMCID: PMC11911097 DOI: 10.1016/j.jtos.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/07/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
PURPOSE To ascertain the homing of monocytes and neutrophils in the epithelium versus stroma of HSV-1 infected corneas at different stages of infection and functional significance of their anatomical location in virus-infected corneas. METHODS The corneas of C57BL/6J mice were infected with HSV-1 McKrae. Mice were euthanized on different days post-infection. The epithelium and stroma were separated from the infected corneas, and flow cytometry was performed to characterize the myeloid cell subsets in the epithelium versus the stromal layers of an infected cornea. MACS columns were used to purify neutrophils or deplete myeloid cells from infected corneas. Corneal epithelial scratch assay was performed to ascertain the impact of neutrophils on epithelium wound healing. RESULTS Our results showed a biphasic influx of monocytes in the epithelial but not the stromal layer of HSV-1-infected corneas. Furthermore, we noted the predominance of monocytes over neutrophils in the epithelium and the stromal layer of the cornea during the pre-clinical stage of corneal HSV-1 infection. However, neutrophils were the major myeloid cell subset in the epithelium and stroma during the clinical disease period of infection. Removal of monocytes from the infected epithelial layer during the pre-clinical stage promotes the dissemination of the virus. Interestingly, neutrophils localized in the corneal epithelium inhibit corneal epithelial wound healing. CONCLUSIONS Together, our data suggest that differential kinetics of monocytes and neutrophils homing in the epithelial layer regulate viral dissemination and epithelial wound healing in HSV-1-infected corneas.
Collapse
MESH Headings
- Animals
- Neutrophils/physiology
- Neutrophils/pathology
- Herpesvirus 1, Human/physiology
- Mice, Inbred C57BL
- Wound Healing/physiology
- Mice
- Monocytes/pathology
- Monocytes/physiology
- Epithelium, Corneal/virology
- Epithelium, Corneal/pathology
- Epithelium, Corneal/immunology
- Keratitis, Herpetic/virology
- Keratitis, Herpetic/pathology
- Keratitis, Herpetic/immunology
- Disease Models, Animal
- Flow Cytometry
- Eye Infections, Viral/virology
- Eye Infections, Viral/pathology
- Eye Infections, Viral/immunology
- Corneal Stroma/virology
- Corneal Stroma/pathology
Collapse
Affiliation(s)
- Mizumi Setia
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Pratima Krishna Suvas
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mashidur Rana
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Anish Chakraborty
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Susmit Suvas
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
13
|
Gaertner K, Terzioglu M, Michell C, Tapanainen R, Pohjoismäki J, Dufour E, Saari S. Species differences in glycerol-3-phosphate metabolism reveals trade-offs between metabolic adaptations and cell proliferation. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2025; 1866:149530. [PMID: 39631556 DOI: 10.1016/j.bbabio.2024.149530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/05/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The temperate climate-adapted brown hare (Lepus europaeus) and the cold-adapted mountain hare (Lepus timidus) are closely related and interfertile species. However, their skin fibroblasts display distinct gene expression profiles related to fundamental cellular processes. This indicates important metabolic divergence between the two species. Through targeted metabolomics and metabolite tracing, we identified species-specific variations in glycerol 3-phosphate (G3P) metabolism. G3P is a key metabolite of the G3P shuttle, which transfers reducing equivalents from cytosolic NADH to the mitochondrial electron transport chain (ETC), consequently regulating glycolysis, lipid metabolism, and mitochondrial bioenergetics. Alterations in G3P metabolism have been implicated in multiple human pathologies including cancer and diabetes. We observed that mountain hare mitochondria exhibit elevated G3P shuttle activity, alongside increased membrane potential and decreased mitochondrial temperature. Silencing mitochondrial G3P dehydrogenase (GPD2), which couples the conversion of G3P to the ETC, uncovered its species-specific role in controlling mitochondrial membrane potential and highlighted its involvement in skin fibroblast thermogenesis. Unexpectedly, GPD2 silencing enhanced wound healing and cell proliferation rates in a species-specific manner. Our study underscores the pivotal role of the G3P shuttle in mediating physiological, bioenergetic, and metabolic divergence between these hare species.
Collapse
Affiliation(s)
- Kateryna Gaertner
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mügen Terzioglu
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Craig Michell
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland; King Abdullah University of Science and Technology, Makkah, Saudi Arabia
| | - Riikka Tapanainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Jaakko Pohjoismäki
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Eric Dufour
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Sina Saari
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
14
|
Moon S, Ito Y. A simplified in vitro disease-mimicking culture system can determine the angiogenic effect of medicines on vascular diseases. Cytotechnology 2025; 77:75. [PMID: 40062227 PMCID: PMC11889311 DOI: 10.1007/s10616-025-00736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/27/2025] [Indexed: 03/21/2025] Open
Abstract
Many patients undergoing clinical regenerative treatments experience severe conditions arising from endothelial disruption. In chronic cardiac and perivascular diseases, deficiencies in vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF), and heparin, which are essential for maintaining and activating endothelial cells, can lead to angiogenic dysregulation. Endothelial disruption caused by ischemic hypoxia and a deficiency in these factors is associated with many vascular diseases. However, their pathogenic processes remain unclear at the cellular level. Therefore, the present study aimed to develop a culture system that mimics the disease environment to test the effectiveness of drug candidates in restoring damaged blood vessels in chronic vascular diseases, including coronary artery disease and peripheral vascular disease. This study focused on VEGF, IGF, and heparin and developed a pseudo-disease culture system by pre-treating human umbilical vein endothelial cells (HUVECs) with a starvation medium (EGM-2™ medium lacking VEGF, IGF, and heparin) to examine the ability of HUVECs to form a traditional 2D vascular network. The results indicated that a deficiency in these proteins results in disruptions in tube morphogenesis. Moreover, the results suggested that dysregulation of the PI3K/AKT pathway plays a key role for in vascular disruption in HUVECs. The proposed pseudo-disease starvation system provides a simple way to visualize pathological disruptions to blood vessels and assess the efficacy of drugs for vascular regeneration. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00736-4.
Collapse
Affiliation(s)
- SongHo Moon
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki Japan
| | - Yuzuru Ito
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki Japan
- Life Science Development Department, CHIYODA Corporation, Yokohama, Kanagawa Japan
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki Japan
| |
Collapse
|
15
|
Pamulang YV, Oontawee S, Rodprasert W, Padeta I, Sa-Ard-Lam N, Mahanonda R, Osathanon T, Somparn P, Pisitkun T, Torsahakul C, Sawangmake C. Potential upscaling protocol establishment and wound healing bioactivity screening of exosomes isolated from canine adipose-derived mesenchymal stem cells. Sci Rep 2025; 15:10617. [PMID: 40148423 PMCID: PMC11950392 DOI: 10.1038/s41598-025-93219-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Mesenchymal stem cell-derived exosomes exhibit promising potential in tissue regeneration. Recent studies highlight its significant therapeutic potential in various stages of wound healing. However, the clinical translation of exosome-based therapy was hindered due to issues regarding low productivity and the lack of efficient production protocol to obtain a clinically relevant exosome quantity. Therefore, this study established a potential upscaling protocol to produce exosomes derived from canine adipose-derived mesenchymal stem cells (cAD-MSCs) and explored its potential for wound treatment. The potential upscaling protocol, termed VSCBIC-3-3D, was carried out using VSCBIC-3 in-house serum-free exosome-collecting solution in a three-dimensional (3D) culture system followed by the tangential flow filtration (TFF) isolation. Our findings suggest that culturing cAD-MSCs with VSCBIC-3 maintained cell morphology and viability. Compared to conventional two-dimensional (2D) protocols, The potential upscaling protocol increased exosome yield and concentration in conditioned medium by 2.4-fold and 3.2-fold, respectively. The quality assessment revealed enhanced purity and bioactivity of exosomes produced using the VSCBIC-3-3D protocol. In addition, the cAD-MSCs-derived exosomes were shown to significantly improve fibroblast migration, proliferation, and wound healing-related gene expression in vitro. This study collectively demonstrates that potential upscaling protocol establishment allowed robust production of exosomes from cAD-MSCs, which exhibit therapeutic potential for wound healing in vitro.
Collapse
Affiliation(s)
- Yudith Violetta Pamulang
- The International Graduate Program of Veterinary Science and Technology (VST), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Saranyou Oontawee
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Watchareewan Rodprasert
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Irma Padeta
- The International Graduate Program of Veterinary Science and Technology (VST), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Noppadol Sa-Ard-Lam
- Immunology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Periodontal Disease and Dental Implant, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Rangsini Mahanonda
- Immunology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Periodontal Disease and Dental Implant, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Poorichaya Somparn
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chutirat Torsahakul
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chenphop Sawangmake
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok, 10330, Thailand.
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
16
|
Alsheleh T, Zraikat M, Daoud F, Alqudah DA, Abdelghany S, Abu Siniyeh A, Alshaer W. In vitro cellular interaction of drug-loaded liposomes with 2D and 3D cell culture of U87-MG cell line. PLoS One 2025; 20:e0320374. [PMID: 40131912 PMCID: PMC11936199 DOI: 10.1371/journal.pone.0320374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
The distinctive physiological and physical properties of 3D cultures that mimic tumor microenvironments in vivo make them more suitable for assessing the efficacy of drugs and nanoparticles compared to 2D culture models. Therefore, this study aims to examine and contrast how liposomes interact with cell cultures in both 2D and 3D models. Hanging drop technique was used to generate 3D spheroids. Cellular toxicity of Doxorubicin and Doxil®-liposomes was tested using an MTT assay. Cellular uptake of Doxil®-liposomes was investigated in 3D and 2D cell culture models using flow cytometry and confocal microscopy. Finally, migration and invasion assays were used to investigate the Doxil®-liposomes interaction with the two models 2D model and 3D model, respectively. Our findings show that cells were able to form spheroid structures when a specific cell ratio was maintained. Flow cytometry analysis revealed that 2D cells exhibited higher Doxil®-liposome uptake than 3D cells. The data obtained from confocal and fluorescent microscopy supported the findings of the flow cytometry analysis. Furthermore, the MTT assay showed that Doxil®-liposomes induced less metabolic-disruption compared to free Doxorubicin. Our results also demonstrated that Doxil®-liposomes interacted more loosely with the 3D model than 2D cells, which was further confirmed by measurements of the total migration and invasion areas. Therefore, a 3D model replicating the in vivo conditions of tumor structure and extracellular matrix to assess the delivery of liposomal-nanoparticles to spheroids through a collagen matrix can be more informative and recapitulate the in vivo microenvironment than the 2D model.
Collapse
Affiliation(s)
- Tasneem Alsheleh
- Department of Biology, Faculty of Science, The University of Jordan, Amman, Jordan
| | - Manar Zraikat
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Fadwa Daoud
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Dana A. Alqudah
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Sharif Abdelghany
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| | - Ahmed Abu Siniyeh
- Department of Clinical Laboratory Sciences, Faculty of Science, The University of Jordan, Amman, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| |
Collapse
|
17
|
Cui X, Wang Y, Li X, Li H, Yin R, Liu Y, Ma A, Yang S. A Positive Feedback Loop Between CXCL16 and the Inflammatory Factors IL-17A and TGF- β Promotes Large Artery Atherosclerosis by Activating the STAT3/NF- κB Pathway. Cardiovasc Ther 2025; 2025:2973633. [PMID: 40165931 PMCID: PMC11957866 DOI: 10.1155/cdr/2973633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 03/01/2025] [Indexed: 04/02/2025] Open
Abstract
CXC chemokine ligand 16 (CXCL16) expression is often observed in studies related to atherosclerosis (AS). However, the process by which CXCL16 promotes AS is still unknown. CXCL16 has the potential to be a therapeutic target for atherosclerotic disease, and we studied whether CXCL16 expression in carotid atherosclerotic plaques is correlated with plaque stability. The results revealed that the expression level of CXCL16 in unstable plaques was greater than that in stable plaques (p < 0.05). In an in vitro model, CXCL16 promoted the expression of interleukin-17A (IL-17A) and transforming growth factor-β (TGF-β) and the release of STAT3/NF-κB pathway-associated proteins by regulating the expression of IL-17A, TGF-β, and CXCL16. In conclusion, there is a positive feedback regulatory pathway between inflammatory factors and CXCL16 during the progression of carotid AS. Inflammatory factors and CXCL16 promote each other's expression and activate the STAT3/NF-κB pathway to promote carotid AS. CXCL16 is highly expressed in carotid atherosclerotic plaques, affecting plaque stability and further leading to the development of AS-related diseases such as ischaemic stroke. Thus, we hypothesise that CXCL16 is a potential therapeutic target for treating AS and AS-related diseases.
Collapse
Affiliation(s)
- Xuechen Cui
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuan Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuening Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruihua Yin
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yue Liu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aijun Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shaonan Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
18
|
Gaertner K, Tapanainen R, Saari S, Fekete Z, Goffart S, Pohjoismäki JLO, Dufour E. Exploring mitonuclear interactions in the regulation of cell physiology: Insights from interspecies cybrids. Exp Cell Res 2025; 446:114466. [PMID: 39978712 DOI: 10.1016/j.yexcr.2025.114466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Brown hares (Lepus europaeus) and mountain hares (Lepus timidus) frequently hybridize in regions where their range overlaps, producing fertile offspring and enabling gene flow between the species. Despite this, no hybrid species has emerged, suggesting that hybrid backcrosses may incur fitness costs. One potential mechanism for such costs involves the interactions between mitochondrial and nuclear gene products, where incompatibilities between species-specific alleles may reinforce species barriers and lead to hybrid breakdown. However, direct experimental evidence for this hypothesis remains limited. In this study, we used fibroblasts derived from skin biopsies of wild-caught hares to generate cytoplasmic hybrid (cybrid) cell lines, wherein mitochondria and mtDNA from one species were transferred to mitochondria-depleted cells of the other species, creating novel mitonuclear gene combinations while preserving the original diploid nuclear background. Employing a range of techniques - including transcriptomics, metabolomics, microscopy, and respirometry - we explored the consequences of mitochondrial transfer between these hare species. Our results reveal that in the studied species mitonuclear incompatibilities exhibit strong effects on cellular fitness but are limited to specific genotypes. We propose mechanisms of cellular-level incompatibility and their potential consequences for interspecific hybrids, offering new insights into the complexity of mitonuclear interactions.
Collapse
Affiliation(s)
- Kateryna Gaertner
- Faculty of Medicine and Health Technology, FI-33520, Tampere University, Finland
| | - Riikka Tapanainen
- Department of Environmental and Biological Sciences, FI-80101, University of Eastern Finland, Finland
| | - Sina Saari
- Faculty of Medicine and Health Technology, FI-33520, Tampere University, Finland
| | - Zsófia Fekete
- Department of Environmental and Biological Sciences, FI-80101, University of Eastern Finland, Finland
| | - Steffi Goffart
- Department of Environmental and Biological Sciences, FI-80101, University of Eastern Finland, Finland
| | - Jaakko L O Pohjoismäki
- Department of Environmental and Biological Sciences, FI-80101, University of Eastern Finland, Finland.
| | - Eric Dufour
- Faculty of Medicine and Health Technology, FI-33520, Tampere University, Finland.
| |
Collapse
|
19
|
Yu J, Chen L, Wang G, Qian K, Weng H, Yang Z, Zheng H, Lu M. RBPMS inhibits bladder cancer metastasis by downregulating MYC pathway through alternative splicing of ANKRD10. Commun Biol 2025; 8:367. [PMID: 40044952 PMCID: PMC11882939 DOI: 10.1038/s42003-025-07842-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/27/2025] [Indexed: 03/09/2025] Open
Abstract
RNA-binding proteins (RBPs) are pivotal mediators of the alternative splicing (AS) machinery of pre-mRNA. Research has demonstrated that the AS process is significantly dysregulated and plays a crucial role in bladder cancer (BLCA). We conducted comprehensive screening and analysis of the TCGA-BLCA cohort, specifically focusing on genes with significant differences in expression levels between carcinoma and adjacent non-cancerous tissues. Among the 500 differentially expressed genes, 5 RNA-binding proteins were identified. Only the RNA-binding protein with multiple splicing (RBPMS) demonstrated a consistent downregulation in BLCA and was correlated with an unfavorable prognosis for affected patients. Subsequent experiments revealed that RBPMS exerted inhibitory effects on the epithelial-mesenchymal transition (EMT) pathway and the migratory potential of BLCA cells. RNA-Seq analysis identified ANKRD10 as a key target mRNA regulated by RBPMS in BLCA. RBPMS depletion in BLCA cells resulted in AS of ANKRD10 and increased ANKRD10-2 expression. ANKRD10-2 functioned as a transcriptional co-activator of MYC proteins, thereby augmenting their transcriptional activity. Furthermore, ANKRD10-2 knockdown significantly rescued the migration enhancement induced by RBPMS depletion in BLCA cells. Taken together, this study revealed a mechanism whereby RBPMS suppresses the migration and invasion of BLCA cells by attenuating MYC pathway activity via the AS of ANKRD10.
Collapse
Affiliation(s)
- Jingtian Yu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Urological Diseases, Wuhan, China
- Hubei Clinical Research Center for Laparoscopic/Endoscopic Urologic Surgery, Wuhan, China
| | - Liang Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Urological Diseases, Wuhan, China
- Institute of Urology, Wuhan University, Wuhan, China
| | - Gang Wang
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kaiyu Qian
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hong Weng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhonghua Yang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Wuhan Clinical Research Center for Urogenital Tumors, Wuhan, China
| | - Hang Zheng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Clinical Research Center for Laparoscopic/Endoscopic Urologic Surgery, Wuhan, China.
- Wuhan Clinical Research Center for Urogenital Tumors, Wuhan, China.
| | - Mengxin Lu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Urological Diseases, Wuhan, China.
| |
Collapse
|
20
|
Robinson KS, Sennhenn P, Yuan DS, Liu H, Taddei D, Qian Y, Luo W. TMBIM6/BI-1 is an intracellular environmental regulator that induces paraptosis in cancer via ROS and Calcium-activated ERAD II pathways. Oncogene 2025; 44:494-512. [PMID: 39609612 PMCID: PMC11832424 DOI: 10.1038/s41388-024-03222-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024]
Abstract
Transmembrane B cell lymphoma 2-associated X protein inhibitor motif-containing (TMBIM) 6, also known as Bax Inhibitor-1 (BI-1), has been heavily researched for its cytoprotective functions. TMBIM6 functional diversity includes modulating cell survival, stress, metabolism, cytoskeletal dynamics, organelle function, regulating cytosolic acidification, calcium, and reactive oxygen species (ROS). Clinical research shows TMBIM6 plays a key role in many of the world's top diseases/injuries (i.e., Alzheimer's, Parkinson's, diabetes, obesity, brain injury, liver disease, heart disease, aging, etc.), including cancer, where TMBIM6 expression impacts patient survival, chemoresistance, cancer progression, and metastasis. We show TMBIM6 is activated by, and undergoes, different conformational changes that dictate its function following a significant change in the cell's IntraCellular Environment (ICE). TMBIM6 agonism, following ICE change, can help the cell overcome multiple stresses including toxin exposure, viral infection, wound healing, and excitotoxicity. However, in cancer cells TMBIM6 agonism results in rapid paraptotic induction irrespective of the cancer type, sub-type, genotype or phenotype. Furthermore, the level of TMBIM6 expression in cancer did not dictate the level of paraptotic induction; however, it did dictate the rate at which paraptosis occurred. TMBIM6 agonism did not induce paraptosis in cancer via canonical routes involving p38 MAPK, JNK, ERK, UPR, autophagy, proteasomes, or Caspase-9. Instead, TMBIM6 agonism in cancer upregulates cytosolic Ca2+ and ROS, activates lysosome biogenesis, and induces paraptosis via ERAD II mechanisms. In xenograft models, we show TMBIM6 agonism induces rapid cancer cell death with no toxicity, even at high doses of TMBIM6 agonist (>450 mg/kg). In summary, this study shows TMBIM6's functional diversity is only activated by severe ICE change in diseased/injured cells, highlighting its transformative potential as a therapeutic target across various diseases and injuries, including cancer.
Collapse
Affiliation(s)
| | | | | | - Hai Liu
- Viva Biotech, Shanghai, China
| | | | | | - Wei Luo
- MicroQuin, Cambridge, MA, USA
| |
Collapse
|
21
|
Poorkazem H, Saber M, Moradmand A, Yakhkeshi S, Seydi H, Hajizadeh-Saffar E, Shekari F, Hassani SN. Comparative effects of various extracellular vesicle subpopulations derived from clonal mesenchymal stromal cells on cultured fibroblasts in wound healing-related process. Int J Biochem Cell Biol 2025; 180:106737. [PMID: 39828140 DOI: 10.1016/j.biocel.2025.106737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/25/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Non-healing wounds pose significant challenges and require effective therapeutic interventions. Extracellular vesicles (EVs) have emerged as promising cell-free therapeutic agents in tissue regeneration. However, the functional differences between different subpopulations of EVs in wound healing remain understudied. This study aimed to evaluate the effects of two distinct subpopulations of clonal mesenchymal stromal cells (cMSC)-derived EVs (cMSC-EVs), namely 20 K and 110K-cMSC-EVs, primarily on in vitro wound healing process, providing fast and cost-effective alternatives to animal models. METHODS In vitro assays were conducted to compare the effects of 20 K and 110K-cMSC-EVs, isolated through high-speed centrifugation and differential ultracentrifugation, respectively. For evaluation the main mechanisms of wound healing, including cell proliferation, cell migration, angiogenesis, and contraction. Human dermal fibroblasts (HDF) were considered as the main cells for analysis of these procedures. Moreover, gene expression analysis was performed to assess the impact of these EV subpopulations on the related process of wound healing on HDF. RESULTS The results demonstrated that both 20 K and 110K-cMSC-EVs exhibited beneficial effects on cell proliferation, cell migration, angiogenesis, and gel contraction. RT-qPCR revealed that both EV types downregulated interleukin 6 (IL6), induced proliferation by upregulating proliferating cell nuclear antigen (PCNA), and regulated remodeling by upregulating matrix metallopeptidase 1 (MMP1) and downregulating collagen type 1 (COL1). DISCUSSION This study highlights the effects of both 20 K and 110K-cMSC-EVs on the potency of HDFs in wound healing-related process. As the notable finding, 20K-cMSC-EVs offer a more feasible and cost-effective subpopulation for isolation and follow the GMP standard, recommended to utilize this fraction for therapeutic application.
Collapse
Affiliation(s)
- Hedie Poorkazem
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Maryam Saber
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Azadeh Moradmand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Yakhkeshi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Homeyra Seydi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
22
|
Ahmad R, Luka M, Journe A, Gallet S, Hegron A, Do Cruzeiro M, Millan MJ, Delagrange P, Masri B, Dam J, Prevot V, Jockers R. Orphan GPR50 Restrains Neurite Outgrowth and Cell Migration by Activating the G 12/13 Protein-RhoA Pathway in Neural Progenitor Cells and Tanycytes. J Pineal Res 2025; 77:e70041. [PMID: 40091563 PMCID: PMC11911906 DOI: 10.1111/jpi.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Human genetic variants of the orphan G protein-coupled receptor GPR50 are suggested risk factors for neuropsychiatric disorders. However, the function of GPR50 in the central nervous system (CNS) and its link to CNS disorders remain poorly defined. Here, we generated GPR50 knockout (GPR50-KO) mice and show that the absence of GPR50 increases neurite outgrowth, cell motility and migration of isolated neural progenitor cells (NPCs) and hypothalamic radial glial cells (tanycytes). These observations were phenocopied in NPCs and tanycytes from wild-type mice treated with neutralizing antibodies the against the prototypical neurite growth inhibitor Nogo-A. Treatment of NPCs and tanycytes from GPR50-KO cells with neutralizing antibodies had no further, additive, effect. Inhibition of neurite growth by GPR50 occurs through activation of the G12/13 protein-RhoA pathway in a manner similar to, but independent of Nogo-A and its receptors. Collectively, we show that GPR50 acts as an inhibitor of neurite growth and cell migration in the brain by activating the G12/13 protein-RhoA pathway.
Collapse
Affiliation(s)
- Raise Ahmad
- Université Paris Cité, Institut Cochin, INSERM, CNRSParisFrance
| | - Marine Luka
- Université Paris Cité, Institut Cochin, INSERM, CNRSParisFrance
| | | | - Sarah Gallet
- University Lille, Inserm, CHU Lille, Lille Neuroscience and CognitionLilleFrance
| | - Alan Hegron
- Université Paris Cité, Institut Cochin, INSERM, CNRSParisFrance
| | | | | | | | - Bernard Masri
- Université Paris Cité, Institut Cochin, INSERM, CNRSParisFrance
| | - Julie Dam
- Université Paris Cité, Institut Cochin, INSERM, CNRSParisFrance
| | - Vincent Prevot
- University Lille, Inserm, CHU Lille, Lille Neuroscience and CognitionLilleFrance
| | - Ralf Jockers
- Université Paris Cité, Institut Cochin, INSERM, CNRSParisFrance
| |
Collapse
|
23
|
Ribeiro A, Pereira-Leite C, Rosado C, Aruci E, Colley HE, Kortekaas Krohn I, Baldea I, Pantelić I, Fluhr JW, Simões SI, Savić S, Costa Lima SA. Enhancing Transcutaneous Drug Delivery: Advanced Perspectives on Skin Models. JID INNOVATIONS 2025; 5:100340. [PMID: 39925780 PMCID: PMC11803873 DOI: 10.1016/j.xjidi.2024.100340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 02/11/2025] Open
Abstract
Skin acts as a dynamic interface with the environment. Pathological alterations in the skin barrier are associated with skin diseases. These conditions are characterized by specific impairments in epidermal barrier functions. Despite its protective nature, the skin can be a relevant route of drug administration, both for topical and transdermal therapy, allowing for improved drug delivery and reducing the incidence of adverse reactions. This manuscript reviews transcutaneous drug delivery as a strategy for treating localized and systemic conditions, highlighting the importance of skin models in the evaluation of drug efficacy and barrier function. It explores advances in in vitro, ex vivo, in vivo, and in silico models for studying cellular uptake, wound healing, oxidative stress, anti-inflammatory, and immune modulation activities. Disease-specific skin models are also discussed.
Collapse
Affiliation(s)
- Ana Ribeiro
- Nanosafety Group, International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Catarina Pereira-Leite
- CBIOS - Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Lisboa, Portugal
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Catarina Rosado
- CBIOS - Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Lisboa, Portugal
| | - Edlira Aruci
- Research Center for the Study of Rare Diseases, Western Balkans University, Tirana, Albania
| | - Helen E. Colley
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Inge Kortekaas Krohn
- Skin Immunology & Immune Tolerance (SKIN) Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Dermatology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Ioana Baldea
- Department of Physiology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ivana Pantelić
- Department of Pharmaceutical Technology and Cosmetology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Joachim W. Fluhr
- Institute of Allergology IFA and Fraunhofer ITMP Immunology and Allergology IA, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra I. Simões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Snežana Savić
- Department of Pharmaceutical Technology and Cosmetology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Sofia A. Costa Lima
- LAQV, REQUIMTE, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
24
|
Suchithra KV, Hameed A, Surya S, Mahammad S, Arun AB. Dual phage-incorporated electrospun polyvinyl alcohol-eudragit nanofiber matrix for rapid healing of diabetic wound infected by Pseudomonas aeruginosa and Staphylococcus aureus. Drug Deliv Transl Res 2025; 15:1092-1108. [PMID: 38980574 DOI: 10.1007/s13346-024-01660-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
Diabetic wound healing remains a healthcare challenge due to co-occurring multidrug-resistant (MDR) bacterial infections and the constraints associated with sustained drug delivery. Here, we integrate two new species of phages designated as PseuPha1 and RuSa1 respectively lysing multiple clinical MDR strains of P. aeruginosa and S. aureus into a novel polyvinyl alcohol-eudragit (PVA-EU†) nanofiber matrix through electrospinning for rapid diabetic wound healing. PVA-EU† evaluated for characteristic changes that occurred due to electrospinning and subjected to elution, stability and antibacterial assays. The biocompatibility and wound healing ability of PVA-EU† were assessed through mouse fibroblast cell line NIH3T3, followed by validation through diabetic mice excision wound co-infected with P. aeruginosa and S. aureus. The electrospinning resulted in the incorporation of ~ 75% active phages at PVA-EU†, which were stable at 25 °C for 30 days and at 4 °C for 90 days. PVA-EU† showed sustained release of phages for 18 h and confirmed to be detrimental to both mono- and mixed-cultures of target pathogens. The antibacterial activity of PVA-EU† remained unaltered in the presence of high amounts of glucose, whereas alkaline pH promoted the activity. The matrix exerted no cytotoxicity on NIH3T3, but showed significant (p < 0.0001) wound healing in vitro and the process was rapid as validated through a diabetic mice model. The sustained release, quick wound closure, declined abundance of target MDR bacteria in situ and histopathological signs of recovery corroborated the therapeutic efficacy of PVA-EU†. Taken together, our data signify the potential application of PVA-EU† in the rapid treatment of diabetic wounds without the aid of antibiotics.
Collapse
Affiliation(s)
- Kokkarambath Vannadil Suchithra
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India
| | - Asif Hameed
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India.
| | - Suprith Surya
- Advanced Surgical Skill Enhancement Division (ASSEND), Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India
| | - Sajida Mahammad
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India
| | - Ananthapadmanabha Bhagwath Arun
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore-575018, India.
- Yenepoya Institute of Arts, Science, Commerce and Management, Balmatta, Mangalore-575002, India.
| |
Collapse
|
25
|
Liu W, Lu D, Jia S, Yang Y, Meng F, Du Y, Yang Y, Yuan L, Nan Y. Molecular mechanism of Gancao Xiexin Decoction regulating EMT and suppressing hepatic metastasis of gastric cancer via the TGF-β1/SMAD pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119430. [PMID: 39900270 DOI: 10.1016/j.jep.2025.119430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gastric cancer (GC) is a highly malignant tumor of the digestive tract, posing a significant menace to human health. Gancao Xiexin Decoction (GCXXD), being a traditional Chinese medicine (TCM), has a good effect on inhibiting the proliferation and metastasis of GC. However, its mechanisms still need further investigation. AIM OF STUDY To investigate the mechanism by which GCXXD inhibits GC metastasis through network pharmacology, and to verify through in vivo and in vitro experiments. MATERIALS AND METHODS The TCMSP and GEO databases, in combination with UPLC-MS/MS techniques, were employed to identify the hub genes, active ingredients, and critical pathways of GCXXD in the treatment of GC. Subsequently, molecular docking was conducted on both the hub genes and the core components. Finally, based on the results of the bioinformatics analysis, the role of GCXXD in inhibiting liver metastasis of GC was elucidated through in vivo and in vitro experiments, including scratch assays, Transwell assays, HE staining, immunohistochemistry, in vivo live imaging, qRT-PCR, and Western blotting. RESULTS Utilizing UPLC-MS/MS and network pharmacology, we identified 20 active ingredients and 5 hub targets in the treatment of GC by GCXXD. Through KEGG analyses, GCXXD treatment of GC could through the TGF-beta pathway. In vivo and in vitro experiments, GCXXD downregulated the mRNA and protein expression level of hub genes involved in the TGF-β1/SMAD pathway and the EMT process. Additionally, GCXXD significantly reduced the incidence of liver metastases in GC. CONCLUSION GCXXD inhibited EMT via blocking the TGF-β1/SMAD pathway, which suppressed GC cell growth and liver metastasis. This study provides data to support the treatment of liver metastasis in GC with TCM and holds significant importance for the research and development of new anticancer drugs.
Collapse
Affiliation(s)
- Wenjing Liu
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Doudou Lu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Shumin Jia
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yating Yang
- The Second Hospital of Chinese Medicine of BAO JI City, Baoji, 721300, Xian, China
| | - Fandi Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Yang
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Nan
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
26
|
Pattadakal S, Ghatti V, Chapi S, G. V, Kumarswamy YK, Raghu MS, T. VG, Nandihalli N, Kasai DR. Poly(vinyl alcohol) Nanocomposites Reinforced with CuO Nanoparticles Extracted by Ocimum sanctum: Evaluation of Wound-Healing Applications. Polymers (Basel) 2025; 17:400. [PMID: 39940602 PMCID: PMC11820508 DOI: 10.3390/polym17030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
This study focused on the synthesis of plant-mediated copper-oxide nanoparticles (OsCuONPs) via the sol-gel technique and the fabrication of OsCuONP-infused PVA composite films (POsCuONPs) utilizing the solvent casting method for wound-healing applications. The prepared OsCuONPs and nanocomposite films were characterized using UV-visible spectra, FTIR, SEM, XRD, TGA, water contact-angle (WCA) measurements, and a Universal testing machine (UTM) for mechanical property measurements. The UV and FTIR tests showed that OsCuONPs were formed and were present in the PVA composite film. Moreover, the mechanical study confirmed that there is an increase in the tensile strength (TS) and Young's modulus (Ym) with 21.75 MPa to 32.50 MPa for TS and 24.80 MPa to 1128.36 MPa for Ym, and a decrease in the % elongation at break (Eb) (394.32 to 75.6). The TGA and WCA study results demonstrated that PVA films containing OsCuONPs are more stable when subjected to high temperatures and demonstrate a decreased hydrophilicity (60.89° to 89.62°). The cytotoxicity and hemolysis tests showed that the CuONPs-3 containing composite films (PVA/OsCuONPs with a wt. ratio of 1.94/0.06) are safe to use, have a good level of cell viability, and do not break down blood. This is true even at high concentrations. The study also discovered that cells moved considerably in 12 and 24 h (13.12 to 19.26 for OsCuONPs and 312.53 to 20.60 for POsCuONPs), suggesting that 60% of the gaps were filled. Therefore, the fabricated POsCuONP nanocomposites may serve as a promising option for applications in wound healing.
Collapse
Affiliation(s)
- Shrishail Pattadakal
- Department of Chemistry, Faculty of Engineering and Technology, Jain University, Jakkasandra, Bengaluru 562112, Karnataka, India; (S.P.); (V.G.); (Y.K.K.)
| | - Vanita Ghatti
- Department of Chemistry, Faculty of Engineering and Technology, Jain University, Jakkasandra, Bengaluru 562112, Karnataka, India; (S.P.); (V.G.); (Y.K.K.)
| | - Sharanappa Chapi
- Department of Physics, B.M.S. College of Engineering, Bengaluru 560019, Karnataka, India
| | - Vidya G.
- Department of Chemistry, Dayananda Sagar College of Engineering, SM Hills, Kumaraswamy Layout, Bengaluru 560011, Karnataka, India;
| | - Yogesh Kumar Kumarswamy
- Department of Chemistry, Faculty of Engineering and Technology, Jain University, Jakkasandra, Bengaluru 562112, Karnataka, India; (S.P.); (V.G.); (Y.K.K.)
| | - M. S. Raghu
- Department of Chemistry, New Horizon College of Engineering, Bengaluru 560103, Karnataka, India;
| | - Vidyavathi G. T.
- Department of Chemistry, RNS Institute of Technology, Rajarajeshwari Nagar, Bengaluru 560098, Karnataka, India;
| | - Nagaraj Nandihalli
- Critical Materials Innovation Hub, Ames National Laboratory, U.S. Department of Energy, Iowa State University, Ames, IA 50011, USA;
| | - Deepak R. Kasai
- Department of Chemistry, Faculty of Engineering and Technology, Jain University, Jakkasandra, Bengaluru 562112, Karnataka, India; (S.P.); (V.G.); (Y.K.K.)
| |
Collapse
|
27
|
Warrier VP, Venkatachalam S, Sakthivel R, Gromiha MM, Karunagaran D. Combinatorial Effects of 5-Fluorouracil and Menadione on Wnt/β-Catenin Pathway in Human Colorectal Cancer Cells. Appl Biochem Biotechnol 2025; 197:1280-1300. [PMID: 39404999 DOI: 10.1007/s12010-024-05072-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 02/13/2025]
Abstract
The incidence and mortality rates of colorectal cancer (CRC) are alarmingly high, and the scientific community is consistently engaged in developing newer therapeutic options for cancer cure or prevention. The fluoropyrimidine drug, 5-fluorouracil (5FU), remains the first line of treatment against CRC; nevertheless, relapses frequently occur since the cells gain resistance over time through various mechanisms. Studies have highlighted the significance of combinatorial treatment of a Wnt signaling inhibitor and 5FU as a better treatment strategy to overcome 5FU resistance. Small molecules that specifically target and disrupt β-catenin-TCF interaction, a crucial step of the Wnt signaling, are promising in CRC treatment. In this study, we investigated the synergistic cytotoxic activity of menadione with 5FU as the former has previously been shown to downregulate Wnt signaling in CRC cells. Docking and experimental results suggest that the drug combination interfered with key protein-protein interactions in the β-catenin-TCF complex, exerted synergistic anti-cancerous effects in CRC cells, and downregulated the expression of Wnt signaling proteins. Taken together, our data suggest that the simultaneous binding of 5FU and menadione to β-catenin can block Wnt signaling by disrupting β-catenin-TCF interaction and inhibit the proliferation of CRC cells.
Collapse
Affiliation(s)
- Vidya P Warrier
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Sankaran Venkatachalam
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Ramasamy Sakthivel
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India.
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India.
| |
Collapse
|
28
|
Spitalny L, Falco N, England W, Allred T, Spitale RC. Novel photocrosslinking chemical probes utilized for high-resolution spatial transcriptomics. RSC Chem Biol 2025:d4cb00262h. [PMID: 39845105 PMCID: PMC11748054 DOI: 10.1039/d4cb00262h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/26/2024] [Indexed: 01/24/2025] Open
Abstract
The architecture of cells and the tissue they form within multicellular organisms are highly complex and dynamic. Cells optimize their function within tissue microenvironments by expressing specific subsets of RNAs. Advances in cell tagging methods enable spatial understanding of RNA expression when merged with transcriptomics. However, these techniques are currently limited by the spatial resolution of the tagging, the number of RNAs that can be sequenced, and multiplexing to isolate spatially-distinct cells within the same tissue landscape. To address these limitations, we developed CrossSeq, which employs photocrosslinking fluorescent probes and confocal microscopy activation to demarcate user-defined regions of interest on fixed cells for multiplexed spatial transcriptomic analysis. We investigate phenyl azide and diazirine crosslinking scaffolds and define their photoactivity profiles. We then deploy the aryl azide scaffold with three fluorophores for multiplexing on glyoxal fixed cells and analyze the defined populations using flow cytometry. Finally, we apply CrossSeq to investigate an in vitro MDA-MB-231-LM2 metastatic cancer migration model to evaluate changes in gene expression at the migratory cell front versus the exterior population. We anticipate this new technology will be a valuable tool addition as it will enable easier access to spatial transcriptomic analysis for the scientific community using conventional microscopy and analysis techniques.
Collapse
Affiliation(s)
- Leslie Spitalny
- Department of Pharmaceutical Sciences, University of California Irvine California 92697 USA
| | - Natalie Falco
- Department of Pharmaceutical Sciences, University of California Irvine California 92697 USA
| | - Whitney England
- Department of Pharmaceutical Sciences, University of California Irvine California 92697 USA
| | - Tyler Allred
- Department of Pharmaceutical Sciences, University of California Irvine California 92697 USA
| | - Robert C Spitale
- Department of Pharmaceutical Sciences, University of California Irvine California 92697 USA
- Department of Chemistry, University of California Irvine California 92697 USA
- Department of Molecular Biology & Biochemistry, University of California Irvine California 92697 USA
| |
Collapse
|
29
|
Fitriani N, Wilar G, Narsa AC, Elamin KM, Wathoni N. Alginate-Based Hydrogels with Amniotic Membrane Stem Cells for Wound Dressing Application. Stem Cells Cloning 2025; 18:1-13. [PMID: 39816853 PMCID: PMC11730520 DOI: 10.2147/sccaa.s493125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/07/2024] [Indexed: 01/18/2025] Open
Abstract
Objective Chronic wounds are a common clinical problem that necessitate the exploration of novel regenerative therapies. We report a method to investigate the in vitro wound healing capacity of an innovative biomaterial, which is based on amniotic membrane-derived stem cells (AMSCs) embedded in an alginate hydrogel matrix. The aim of this study was to prepare an sodium alginate-based hydrogel, cross-linked calcium chloride (CaCl2) with the active ingredient AMSC (AMSC/Alg-H) and to evaluate its in vitro effectiveness for wound closure. Methods This hydrogel preparation involved combining sterile solutions of AMSC, sodium alginate, and CaCl2, followed by rinsing with serum-free media. The cells were cultured in different 6-well plates, namely sodium alginate, calcium chloride, AMSC, Alg-H, and AMSC/Alg-H, in complete medium with 10% FBS. The hydrogel was successfully formulated, as confirmed by characterization techniques including Scanning Electron Microscopy (SEM), Fourier Transform Infrared (FTIR) spectroscopy, Differential Scanning Calorimetry (DSC), Cytotoxicity Studies, TGF-β1 Level Measurement by ELISA, and Cell Scratch Wound Assay. Results Cryo-EM characterization of the Alg-H preparation successfully demonstrated the encapsulation of MSCs. FTIR and DSC analyses indicate that crosslinking transpires in Alg-H encapsulating AMSC. The AMSC/Alg-H preparation showed no significant difference in toxicity compared to HaCaT cells (p < 0.05), indicating it was not toxic to HaCaT cells. Furthermore, in the scratch wound assay test at 24 hours, the AMSC/Alg-H preparation achieved 100% wound closure, outperforming both AMSC and Alg-H alone. In vitro assessment revealed that AMSC/Alg-H significantly enhanced key wound healing processes, including cell proliferation and migration, compared to Alg-H. Conclusion Our study demonstrated the promising potential of AMSC/Alg-H as an enhanced regenerative therapy for in vitro wound healing. AMSC/Alg-H was able to maintain the viability of AMSCs and facilitate the formation of tissue-like structures.
Collapse
Affiliation(s)
- Nurul Fitriani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
- Pharmaceutical Research and Development Laboratory of FARMAKA TROPIS, Faculty of Pharmacy, Universitas Mulawarman, Samarinda, 75119, Indonesia
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Angga Cipta Narsa
- Pharmaceutical Research and Development Laboratory of FARMAKA TROPIS, Faculty of Pharmacy, Universitas Mulawarman, Samarinda, 75119, Indonesia
| | - Khaled M Elamin
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| |
Collapse
|
30
|
Reddy TS, Privér SH, Ojha R, Mirzadeh N, Velma GR, Jakku R, Hosseinnejad T, Luwor R, Ramakrishna S, Wlodkowic D, Plebanski M, Bhargava SK. Gold(I) complexes of the type [AuL{κC-2-C 6H 4P(S)Ph 2}] [L = PTA, PPh 3, PPh 2(C 6H 4-3-SO 3Na) and PPh 2(2-py)]: Synthesis, characterisation, crystal structures, and In Vitro and In Vivo anticancer properties. Eur J Med Chem 2025; 281:117007. [PMID: 39500067 DOI: 10.1016/j.ejmech.2024.117007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/17/2024] [Accepted: 10/27/2024] [Indexed: 12/02/2024]
Abstract
Four new mononuclear gold (I) compounds of the type [AuL{κC-2-C6H4P(S)Ph2}] {L = PTA (1), PPh3 (2), PPh2(C6H4-3-SO3Na) (3), and PPh2(2-py) (4)} were prepared by scission of the dinuclear compound [Au2{μ-2-C6H4P(S)Ph2}2] by L or via a transmetalation reaction using the organotin reagent 2-Me3SnC6H4P(S)Ph2 and a suitable gold halide precursor. The cytotoxic potential of complexes 1-4 was evaluated against four human cancer cell lines of diverse cellular origin: cervical (HeLa), prostate (PC-3), non-small cell lung adenocarcinoma (A549), and fibrosarcoma (HT-1080). The in vitro cytotoxicity results showed that 1 demonstrated exceptional anticancer activity with IC50 values ranging from 0.08 to 3.5 μM. Complex 3, which contains a sulfonated triphenyl phosphine ligand, displayed the weakest anticancer activity with IC50 values ranging from 3.1 to >50 μM. When compared to the standard chemotherapeutic drug cisplatin, 1 displayed approximately 27-fold greater cytotoxic activity against cervical cancer cells and 3.5- and 7.5-fold greater activities against prostate and fibrosarcoma cancer cells, respectively. Additionally, 1 exhibited 3-fold selectivity for cervical cancer cells compared to non-cancerous HEK-293 cells. Mechanistic investigations revealed that 1 induced apoptosis, which was associated with elevated reactive oxygen species (ROS) and inhibition of the intracellular enzyme thioredoxin reductase. Furthermore, 1 exhibited notable antiangiogenic characteristics in an in vivo model using transgenic zebrafish Tg(fli1a:EGFP). In vivo studies using mouse xenograft models showed that complex 1 displayed superior inhibition of tumour growth (82 %) compared to the clinical drug cisplatin (29 %). Overall, these results highlight the potential of gold (I) compounds as novel antitumour agents.
Collapse
Affiliation(s)
- T Srinivasa Reddy
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Steven H Privér
- School of Science, STEM College, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Ruchika Ojha
- School of Science, STEM College, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Nedaossadat Mirzadeh
- School of Science, STEM College, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Ganga Reddy Velma
- School of Science, STEM College, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Ranjithkumar Jakku
- School of Science, STEM College, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Tayebeh Hosseinnejad
- School of Science, STEM College, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Rodney Luwor
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, 3050, Australia
| | - Sistla Ramakrishna
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Donald Wlodkowic
- The Neurotox Laboratory, School of Science, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Victoria, 3083, Australia.
| | - Suresh K Bhargava
- School of Science, STEM College, RMIT University, Melbourne, Victoria, 3001, Australia.
| |
Collapse
|
31
|
Chen Y, Shen J, Yuan M, Li H, Li Y, Zheng S, Han B, Zhang C, Liu S, Sun Q, Wu J. Dehydrocostus lactone suppresses gastric cancer progression by targeting ACLY to inhibit fatty acid synthesis and autophagic flux. J Adv Res 2025; 67:331-348. [PMID: 38295877 PMCID: PMC11725148 DOI: 10.1016/j.jare.2024.01.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/28/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024] Open
Abstract
INTRODUCTION Dehydrocostus lactone (Dehy), a natural sesquiterpene lactone from Saussurea lappa Clarke, displays remarkable efficacy in treating cancer and gastrointestinal disorders. However, its anti-gastric cancer (GC) effect remains poorly understood. OBJECTIVES Our study aimed to elucidate the anti-GC effect of Dehy and its putative mechanism. METHODS The anti-GC effect was assessed with MTT, colony formation, wound healing and transwell invasion assays. Cell apoptosis rate was detected by Annexin V-FITC/PI binding assay. Network pharmacology analysis and XF substrate oxidation stress test explored the underlying mechanism and altered metabolic phenotype. Lipogenic enzyme expressions and neutral lipid pool were measured to evaluate cellular lipid synthesis and storage. Biolayer interferometry and molecular docking investigated the direct target of Dehy. Autophagosomes were observed by transmission electron microscopy and MDC staining, while the autophagic flux was detected by mRFP-GFP-LC3 transfection. The clinical significance of ACLY was confirmed by tissue microarrays. Patient-derived xenograft (PDX) models were adopted to detect the clinical therapeutic potential of Dehy. RESULTS Dehy prominently suppressed GC progression both in vitro and in vivo. Mechanistically, Dehy down-regulated the lipogenic enzyme ACLY, thereby reducing fatty acid synthesis and lipid reservation. Moreover, IKKβ was identified as the direct target of Dehy. Dehy inhibited the phosphorylation of IKKβ, promoting the ubiquitination and degradation of ACLY, thereby resulting in lipid depletion. Subsequently, GC cells initiated autophagy to replenish the missing lipids, whereas Dehy impeded this cytoprotective mechanism by down-regulating LAMP1 and LAMP2 expressions, which disrupted lysosomal membrane functions, ultimately leading to apoptosis. Additionally, Dehy exhibited potential in GC clinical therapy as it enhanced the efficacy of 5-Fluorouracil in PDX models. CONCLUSIONS Our work identified Dehy as a desirable agent for blunting abnormal lipid metabolism and highlighted its inhibitory effect on protective autophagy, suggesting the future development of Dehy as a novel therapeutic drug for GC.
Collapse
Affiliation(s)
- Yuxuan Chen
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Junyu Shen
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Mengyun Yuan
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Huaizhi Li
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Yaqi Li
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Shanshan Zheng
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Bo Han
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Cancan Zhang
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Shenlin Liu
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China.
| | - Qingmin Sun
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China.
| | - Jian Wu
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
32
|
Agger AE, Samara A, Geng T, Olstad OK, Reseland JE. Mimicking and in vitro validating chronic inflammation in human gingival fibroblasts. Arch Oral Biol 2025; 169:106113. [PMID: 39447377 DOI: 10.1016/j.archoralbio.2024.106113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE The aim of this study was to identify and validate in vitro conditions that may mimic the translational, cytokine and chemokine profiles observed in human inflamed gingiva in vivo. DESIGN Primary human gingiva fibroblast cells (HFIB-G) were cultured under serum starvation conditions (0 - 10 %), supplemented with increasing lipopolysaccharide (LPS) concentrations (0.1, 1, or 10 µg/ml) from two bacterial strains E. coli and P. gingivalis and 0.1, 1, or 10 ng/ml recombinant interleukin 1β (IL-1β), alone or in combinations. The levels of cytokines/chemokines were measured in the cell culture medium by Luminex, and gene expression was quantified by Affymetrix microarrays at 24, 48 and 72 h. RESULTS Inflammation markers were not elevated after stimulation with P. gingivalis LPS, while E. coli LPS and IL-1β individually increased the secretion of interleukin 6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) to the cell culture medium. IL-1β administration also increased the secretion of several factors, including tumor necrosis factor (TNFα). However, the combination of 1 µg/ml E. coli LPS, 1 ng/ml IL-1β and serum starvation led to increased secretion of IL-6, TNFα, in addition to other factors found in inflamed tissue. Gene expression analyses revealed that this combination not only enhanced the expression interleukins/chemokines genes but also T helper cell signaling and matrix metalloproteinases. CONCLUSION Serum reduction in cell culture medium together with the administration of E. coli LPS and IL-1β resulted in gene expression and secreted cytokine/chemokine profiles similar to that found in vivo during chronic inflammation.
Collapse
Affiliation(s)
- Anne Eriksson Agger
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway; FUTURE, Center for Functional Tissue Reconstruction, University of Oslo, Oslo, Norway.
| | - Athina Samara
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway; FUTURE, Center for Functional Tissue Reconstruction, University of Oslo, Oslo, Norway.
| | - Tianxiang Geng
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway.
| | | | - Janne Elin Reseland
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway; FUTURE, Center for Functional Tissue Reconstruction, University of Oslo, Oslo, Norway.
| |
Collapse
|
33
|
Hao X, Chen Y, Sahu D, Przanowska RK, Weidmann CA, Nardi I, Weeks KM, Dutta A. A 36-base hairpin within lncRNA DRAIC , modulated by alternative splicing, interacts with the IKKα coiled-coil domain and inhibits NF-κB and tumor cell phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.23.629241. [PMID: 39764029 PMCID: PMC11703201 DOI: 10.1101/2024.12.23.629241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
The 1.7 kb DRAIC long noncoding RNA inhibits tumor growth, inhibits cancer cell invasion, migration, colony formation and interacts with IKK (IκB kinase) subunits, inhibiting the phosphorylation and degradation of the NF-κB inhibitor, IκB, to suppress the activation of NF-κB. Whether these activities are all linked is unclear. We used SHAPE-MaP to obtain the secondary structure of the lncRNA to perform structure-functions studies which identified the minimal region of DRAIC necessary for repressing NF-κB. A 36-nucleotide hairpin (A+B) within DRAIC inhibits NF-κB, inhibits IκB phosphorylation and binds specifically with the IKKα coiled- coil domain with a very high affinity: K D of ∼1-5 nM. This interaction weakens the dimerization of of the coiled coil domains of two IKK subunits, a dimerization that is indispensable for IKK activity. A+B is sufficient and necessary to inhibit the oncogenic phenotypes in multiple cancer cell-lines, demonstrating that interaction with IKK and inhibition of NF-κB is key for cancer suppression by DRAIC. Presence of this critical hairpin is modulated by alternative splicing the extends exon 4 to exon 4a of DRAIC and the expression of exon 4a in lung tumors is associated with low NF-κB activity. This is also the first demonstration that a short RNA can disrupt coiled- coil dimerization.
Collapse
|
34
|
Amonchaiyapitak O, Santiwong B, Wasanapiarnpong T, Chanamuangkon T, Linsuwanont P. Investigation of the physical, chemical, and biological properties of the cockle shell-derived calcium silicate-based pulp capping material: a pilot study. BDJ Open 2024; 10:93. [PMID: 39681553 DOI: 10.1038/s41405-024-00281-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
INTRODUCTION Hard-setting calcium hydroxide-based materials, e.g., Dycal and Life, have been widely used for direct pulp capping. However, various studies have shown undesirable effects such as high solubility and unpredictable dentine bridge formation. Bioceramic, mainly composed of tricalcium and dicalcium silicates, e.g., mineral trioxide aggregate and Biodentine, have provided more desirable physical and biological properties. This study aims to measure the physical properties, chemical properties, and biological response of human dental pulp cells (HDPCs) on three dental pulp-capping materials, Dycal, Life, and cockle shell-derived tricalcium silicate pulp capping material (C-Cap). METHODS C-Cap was prepared from cockle shells and rice husk ash. Its chemical composition was identified using X-ray diffractometry. The setting time, flow, solubility, and radiopacity tests were performed following the International Organization for Standardization 6876:2012. pH and calcium ion release were measured. The materials were subjected to an extraction medium at various concentrations and subsequently measured for cytotoxicity and migration on HDPCs, from three healthy, mature permanent teeth from different donors. Osteogenic differentiation was assessed by examining alkaline phosphatase enzyme activity and alizarin red staining assay. The data were tested for a normal distribution. The differences among groups were statistically analyzed using ANOVA and Tukey's multiple comparison test (p < 0.05). RESULTS The setting time of each material was approximately 1-2 min. C-Cap showed the lowest solubility (10.27% ± 1.02%) compared to Dycal (12.67% ± 0.94%) and Life (12.74% ± 1.33%), with a significant difference (p < 0.05). All materials exhibited radiopacity ranging from 2.4 to 2.9 mm of aluminum. C-Cap had the highest flow, alkalinity, and calcium ion release. C-Cap was significantly less cytotoxic than Dycal and Life (p < 0.05). The migration of HDPCs cultured in C-Cap extraction medium (27.74% ± 0.12%) was comparable to that in serum-free medium (27.09% ± 0.08%) with a significant difference (p < 0.05). The mineralization by HDPCs maintained in C-Cap extraction medium was significantly higher than those in Dycal and Life extraction mediums with a significant difference (p < 0.05). CONCLUSIONS C-Cap, a tricalcium silicate-based pulp capping material has potential for further development. C-Cap exhibited comparable physical properties and superior biological properties when compared to Dycal and Life.
Collapse
Affiliation(s)
- Orana Amonchaiyapitak
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Wangmai, Pathumwan, Bangkok, 10330, Thailand
| | - Busayarat Santiwong
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Wangmai, Pathumwan, Bangkok, 10330, Thailand
| | - Thanakorn Wasanapiarnpong
- Department of Materials of Science, Faculty of Science, Chulalongkorn University, 254 Phayathai Road, Wangmai, Pathumwan, Bangkok, 10330, Thailand
| | - Theerapat Chanamuangkon
- Biomaterial Testing Center, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Wangmai, Pathumwan, Bangkok, 10330, Thailand
| | - Pairoj Linsuwanont
- Department of Operative Dentistry, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Wangmai, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
35
|
Lara-Hernández G, Ramos-Silva JA, Pérez-Soto E, Figueroa M, Flores-Berrios EP, Sánchez-Chapul L, Andrade-Cabrera JL, Luna-Angulo A, Landa-Solís C, Avilés-Arnaut H. Anticancer Activity of Plant Tocotrienols, Fucoxanthin, Fucoidan, and Polyphenols in Dietary Supplements. Nutrients 2024; 16:4274. [PMID: 39770896 PMCID: PMC11678266 DOI: 10.3390/nu16244274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Plants and algae harbor diverse molecules with antioxidant activity and have been demonstrated to directly inhibit cancer cell growth and mitigate the oxidative damage associated with certain antitumor therapies. While antioxidant supplementation, either alone or in combination with chemotherapy, has shown promise in improving quality of life, further research is needed to explore the effects of antioxidant combinations on specific cancer cell lines. Methods: In this study, the in vitro cytotoxic and apoptotic properties of natural compounds derived from plants and algae, as well as certain dietary supplements, were investigated against various human cancer cell lines, including bone, leukemia, colorectal, breast, and prostate cancers. Results: Apple polyphenols, fucoxanthin, and plant-derived tocotrienols exhibited cytotoxic effects across all lines; however, tocotrienols demonstrated the most potent, time-dependent cytotoxic activity, with a half-inhibitory concentration (IC50) of 4.3 μg/mL in bone cancer cells. Analysis of dietary supplements 2.1, 4.0, and 10.0 revealed that supplement 10.0 exhibited specific cytotoxic activity against bone cancer line TIB-223 and colorectal cancer cell line Caco2, with IC50 values of 126 μg/mL and 158 μg/mL, respectively. Both tocotrienols and supplement 10.0 induced morphological changes in TIB-223 cells, inhibited cell migration (anti-metastatic activity), and promoted apoptosis, as evidenced by caspase 3/7 activation in both bone and colorectal cancer cells. Conclusions: These findings provide valuable insights for the development of targeted dietary supplements to enhance the anticancer effect of conventional chemotherapy in specific cancer types.
Collapse
Affiliation(s)
- Gabriel Lara-Hernández
- Laboratorio de Biomedicina y Salud Ocupacional, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Ciudad de México 07320, Mexico; (G.L.-H.); (E.P.-S.)
| | - José Alberto Ramos-Silva
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Campus Ciudad Universitaria, Av. Universidad S/N, San Nicolás de los Garza 66455, Mexico;
| | - Elvia Pérez-Soto
- Laboratorio de Biomedicina y Salud Ocupacional, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Ciudad de México 07320, Mexico; (G.L.-H.); (E.P.-S.)
| | - Mario Figueroa
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | | | - Laura Sánchez-Chapul
- Laboratorio de Enfermedades Neuromusculares, División de Neurociencias Clínicas, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Ciudad de México 14389, Mexico; (L.S.-C.); (A.L.-A.)
| | - José Luis Andrade-Cabrera
- Laboratorio de Patología Clínica, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Ciudad de México 14389, Mexico;
| | - Alexandra Luna-Angulo
- Laboratorio de Enfermedades Neuromusculares, División de Neurociencias Clínicas, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Ciudad de México 14389, Mexico; (L.S.-C.); (A.L.-A.)
| | - Carlos Landa-Solís
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Ciudad de México 14389, Mexico;
| | - Hamlet Avilés-Arnaut
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Campus Ciudad Universitaria, Av. Universidad S/N, San Nicolás de los Garza 66455, Mexico;
| |
Collapse
|
36
|
Prathibha PM, Thomas NG, Dalvi YB, Varghese KG, Binsi PK, Zynudheen AA, Lekshmi M, Shilpa J, Sajith V, Sukumaran A. Fish scale-derived hydroxyapatite for alveolar ridge preservation. Biotechnol Appl Biochem 2024; 71:1272-1280. [PMID: 38951991 DOI: 10.1002/bab.2627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/01/2024] [Accepted: 05/27/2024] [Indexed: 07/03/2024]
Abstract
Alveolar ridge resorption following tooth extraction poses significant challenges for future dental restorations. This study investigated the efficacy of fish scale-derived hydroxyapatite (FSHA) as a socket preservation graft material to maintain alveolar bone volume and architecture. FSHA was extracted from *Labeo rohita* fish scales and characterized using Fourier transform infrared (FTIR) analysis. In vitro, biocompatibility and osteogenic potential were assessed using Saos-2 human osteosarcoma cells. Cell viability, migration, and proliferation were evaluated using MTT and scratch assays. In vivo performance was assessed in a rat model, and FSHA was compared to a commercial xenograft (Osseograft) and ungrafted controls. Histological analysis was performed at 8-week post-implantation to quantify new bone formation. FTIR confirmed the purity and homogeneity of FSHA. In vitro, FSHA enhanced Saos-2 viability, migration, and proliferation compared to controls. In vivo, FSHA demonstrated superior bone regeneration compared to Osseograft and ungrafted sites, with balanced graft resorption and new bone formation. Histological analysis revealed an active incorporation of FSHA into new bone, with minimal gaps and ongoing remodeling. Approximately 50%-60% of FSHA was resorbed by 8 weeks, closely matching the rate of new bone deposition. FSHA stimulated more bone formation in the apical socket region than in coronal areas. In conclusion, FSHA is a promising biomaterial for alveolar ridge preservation, exhibiting excellent biocompatibility, osteogenic potential, and balanced resorption. Its ability to promote robust bone regeneration highlights its potential as an effective alternative to currently used graft materials in socket preservation procedures.
Collapse
Affiliation(s)
- P M Prathibha
- Department of Oral and Maxillofacial Surgery, Pushpagiri College of Dental Sciences, Pushpagiri Medical Society, Tiruvalla, Kerala, India
| | - N G Thomas
- Department of Periodontology, Pushpagiri College of Dental Sciences, Pushpagiri Medical Society, Tiruvalla, Kerala, India
- Pushpagiri Research Center, Pushpagiri Institute of Medical Sciences and Research Centre, Pushpagiri Medical Society, Tiruvalla, Kerala, India
| | - Y B Dalvi
- Pushpagiri Research Center, Pushpagiri Institute of Medical Sciences and Research Centre, Pushpagiri Medical Society, Tiruvalla, Kerala, India
| | - K G Varghese
- Department of Oral and Maxillofacial Surgery, Pushpagiri College of Dental Sciences, Pushpagiri Medical Society, Tiruvalla, Kerala, India
| | - P K Binsi
- ICAR-Central Institute of Fisheries Technology, Cochin, India
| | - A A Zynudheen
- ICAR-Central Institute of Fisheries Technology, Cochin, India
| | - M Lekshmi
- Department of Periodontology, Pushpagiri College of Dental Sciences, Pushpagiri Medical Society, Tiruvalla, Kerala, India
| | - J Shilpa
- Department of Biotechnology, Sethu Institute of Technology, Virudhunagar, Tamil Nadu, India
| | - Vellappally Sajith
- Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Anil Sukumaran
- Pushpagiri Research Center, Pushpagiri Institute of Medical Sciences and Research Centre, Pushpagiri Medical Society, Tiruvalla, Kerala, India
- Oral Health Institute, Department of Dentistry, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
37
|
Bhoopathy J, Vedakumari Sathyaraj W, Yesudhason BV, Rajendran S, Dharmalingam S, Seetharaman J, Muthu R, Murugesan R, Raghunandhakumar S, Anandasadagopan SK. Haemostatic potency of sodium alginate/aloe vera/sericin composite scaffolds - preparation, characterisation, and evaluation. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:35-45. [PMID: 38112317 DOI: 10.1080/21691401.2023.2293784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023]
Abstract
Fabrication of haemostatic materials with excellent antimicrobial, biocompatible and biodegradable properties remains as a major challenge in the field of medicine. Haemostatic agents play vital role in protecting patients and military individuals during emergency situations. Natural polymers serve as promising materials for fabricating haemostatic compounds due to their efficacy in promoting hemostasis and wound healing. In the present work, sodium alginate/aloe vera/sericin (SA/AV/S) scaffold has been fabricated using a simple cost-effective casting method. The prepared SA/AV/S scaffolds were characterised for their physicochemical properties such as scanning electron microscope, UV-visible spectroscopy and Fourier transform infra-red spectroscopy. SA/AV/S scaffold showed good mechanical strength, swelling behaviour and antibacterial activity. In vitro experiments using erythrocytes proved the hemocompatible and biocompatible features of SA/AV/S scaffold. In vitro blood clotting assay performed using human blood demonstrated the haemostatic and blood absorption properties of SA/AV/S scaffold. Scratch wound assay was performed to study the wound healing efficacy of prepared scaffolds. Chick embryo chorioallantoic membrane assay carried out using fertilised embryos proved the angiogenic property of SA/AV/S scaffold. Thus, SA/AV/S scaffold could serve as a potential haemostatic healthcare product due to its outstanding haemostatic, antimicrobial, hemocompatible, biocompatible and angiogenic properties.
Collapse
Affiliation(s)
- Jayavardhini Bhoopathy
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, Tamil Nadu, India
| | - Weslen Vedakumari Sathyaraj
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, Tamil Nadu, India
| | - Beryl Vedha Yesudhason
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai 600119, India
| | - Selvarajan Rajendran
- Centre for Nano Science and Technology, Alagappa College of Technology Campus, Anna University, Chennai 600025, Tamil Nadu, India
| | - Sankari Dharmalingam
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Jayashri Seetharaman
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Ranjitha Muthu
- Department of Research, Karpaga Vinayaga Institute of Medical Science and Research Centre, GST Road, Chinna Kolambakkam, Palayanoor (PO), Tamil Nadu 603308, Tamil Nadu
| | - Ramachandran Murugesan
- Department of Research, Karpaga Vinayaga Institute of Medical Science and Research Centre, GST Road, Chinna Kolambakkam, Palayanoor (PO), Tamil Nadu 603308, Tamil Nadu
| | | | | |
Collapse
|
38
|
Meloni M, de Rooij B, Janssen FW, Rescigno F, Lombardi B. Targeted Antibacterial Endolysin to Treat Infected Wounds on 3D Full-Thickness Skin Model: XZ.700 Efficacy. Pharmaceutics 2024; 16:1539. [PMID: 39771518 PMCID: PMC11728803 DOI: 10.3390/pharmaceutics16121539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/15/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025] Open
Abstract
Backgrounds/Objectives: Skin wound healing is a physiological process orchestrated by epithelial and mesenchymal cells able to restore tissue continuity by re-organizing themselves and the ECM. This research study aimed to develop an optimized in vitro experimental model of full-thickness skin, to address molecular and morphological modifications occurring in the re-epithelization and wound healing process. Methods: Wound healing starting events were investigated within an experimental window of 8 days at the molecular level by gene expression and immunofluorescence of key epidermal and dermal biomarkers. To mirror the behavior of infected wounds, the established wound healing model was then colonized with S. aureus, and the efficacy of a novel antibacterial agent, XZ.700, was investigated. Viable counts (CFU/tissue), IF, and ultrastructural analysis (SEM) were performed to evaluate S. aureus colonization inside and around the wound bed in an experimental window of 3 h of colonization and 24 h of treatment. Results: Endolysin showed an efficacy in counteracting bacterial growth and invasion within the wound bed, reducing the S. aureus load compared to its placebo, thanks to its selective antimicrobial activity interfering with biofilm formation. Conclusions: The preclinical in vitro infected wound model on FT-kin showed interesting applications to assess the repair efficacy of dermo-pharmaceutical and cosmetic formulations.
Collapse
Affiliation(s)
- Marisa Meloni
- VitroScreen s.r.l., In Vitro Innovation Center, Via Mosè Bianchi 103, 20149 Milan, MI, Italy; (M.M.); (B.L.)
| | - Bob de Rooij
- Micreos Pharmaceuticals, Neuhofstrasse 12, CH-6430 Baar, ZG, Switzerland; (B.d.R.); (F.W.J.)
| | - Ferdinand W. Janssen
- Micreos Pharmaceuticals, Neuhofstrasse 12, CH-6430 Baar, ZG, Switzerland; (B.d.R.); (F.W.J.)
| | - Francesca Rescigno
- VitroScreen s.r.l., In Vitro Innovation Center, Via Mosè Bianchi 103, 20149 Milan, MI, Italy; (M.M.); (B.L.)
| | - Bernadette Lombardi
- VitroScreen s.r.l., In Vitro Innovation Center, Via Mosè Bianchi 103, 20149 Milan, MI, Italy; (M.M.); (B.L.)
| |
Collapse
|
39
|
Sarkar S, Moitra P, Duan W, Bhattacharya S. A Multifunctional Aptamer Decorated Lipid Nanoparticles for the Delivery of EpCAM-targeted CRISPR/Cas9 Plasmid for Efficacious In Vivo Tumor Regression. Adv Healthc Mater 2024; 13:e2402259. [PMID: 39212195 DOI: 10.1002/adhm.202402259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Epithelial cell adhesion molecule (EpCAM) gene encodes a type-I trans-membrane glycoprotein that is overexpressed in many cancerous epithelial cells and promotes tumor progression by regulating the expression of several oncogenes like c-myc and other cyclins. Because of this tumorigenic association, the EpCAM gene has been a potential target for anti-cancer therapy in recent days. Herein, it is attempted to knockout the proto-oncogenic EpCAM expression by efficiently delivering an all-in-one Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) plasmid via a lipid nanoparticle system made out of synthetic stimuli-sensitive lipids. The plasmid possesses the necessary information in the form of a guide RNA targeted to the EpCAM gene. The aptamer decorated system selectively targets EpCAM overexpressed cells and efficiently inhibits the genetic expression. It has explored the pH-responsive property of the developed lipid nanoparticles and monitored their efficacy in various cancer cell lines of different origins with elevated EpCAM levels. The phenomenon has further been validated in vivo in non-immunocompromised mouse tumor models. Overall, the newly developed aptamer decorated lipid nanoparticle system has been proven to be efficacious for the delivery of EpCAM-targeted CRISPR/Cas9 plasmid.
Collapse
Affiliation(s)
- Sourav Sarkar
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India
| | - Parikshit Moitra
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur, Berhampur, Odisha, 760003, India
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Santanu Bhattacharya
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata, 700032, India
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Yerpedu, Tirupati District, Andhra Pradesh, 517619, India
| |
Collapse
|
40
|
Hamada MA, Mohamed ET. Characterization of Serratia marcescens (OK482790)' prodigiosin along with in vitro and in silico validation for its medicinal bioactivities. BMC Microbiol 2024; 24:495. [PMID: 39587466 PMCID: PMC11587630 DOI: 10.1186/s12866-024-03634-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Microbial prodigiosin pigment has been proposed as a promising biomolecule having an antibacterial, immunosuppressive, antimalarial, antineoplastic, and anticancer activities. The good outcome originates from getting natural pigment, which has many medical applications. RESULTS In this investigation, prodigiosin (PG) was extracted, characterized by UV-visible spectroscopy, thin-layer chromatography, mass spectroscopy, Fourier-transform infrared spectroscopy, and tested in various medical applications as an antibacterial, antioxidant, antibiofilm, anticancer, and wound healing agent at different concentrations. Antibacterial activity of PG pigment was shown against both Gram-positive and Gram-negative bacterial strains. Enterococcus faecalis was the most severely impacted, with minimum inhibitory value of 3.9 µg/mL. The formed biofilm by Pseudomonas aeruginosa was suppressed by 58-2.50% at prodigiosin doses ranging from 1000 to 31.25 µg/mL, respectively. The half-maximal inhibitory concentration (IC50) of 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid (ABTS) free radical was 74.18 ± 23.77 µg/mL. At 100 µg/mL concentration, OK482790 prodigiosin had no harmful effect on normal skin cells and exhibited mild wound healing properties. Additionally, molecular docking simulations confirmed the prodigiosin's interactions with target proteins, including epidermal growth factor receptor tyrosine kinase (EGFR-TK, PDB ID: 1M17), peptide deformylase from E. faecalis (PDB ID: 2OS1), acidic fibroblast growth factor (FGF-1, PDB ID: 3K1X), PA14_16140 protein from P. aeruginosa (PDB ID: 8Q8O), and human peroxiredoxin 5 (PDB ID: 1HD2) for explaining the anticancer, antibacterial, wound healing, antibiofilm, and antioxidant activities, respectively. Prodigiosin had favorable binding affinities and putative modes of action across various therapeutic domains. CONCLUSION This study pioneers the use of prodigiosin as a natural alternative to synthetic medicine since it fights germs, heals wounds, is antioxidant, and reduces biofilm formation. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Marwa A Hamada
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt.
| | - Eslam T Mohamed
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
| |
Collapse
|
41
|
Wagner KD, Safwan-Zaiter H, Wagner N. A Dual Role of the Senescence Marker P16Ink4a in Liver Endothelial Cell Function. Cells 2024; 13:1929. [PMID: 39682678 PMCID: PMC11640762 DOI: 10.3390/cells13231929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
P16Ink4a is a well-established marker of senescence. Although P16Ink4a is expressed in endothelial cells, little is known about its function in these cells. Using isolated liver endothelial cells with silencing or overexpression of P16Ink4a, we show here that dependent on P16Ink4a levels, different pathways and functions are affected. High levels of P16Ink4a reduce proliferation and induce senescence, while low levels have the opposite effects. Only high P16Ink4a expression reduces in vitro angiogenesis. Expression profiling reveals an inflammatory phenotype upon silencing of P16Ink4a, while P16Ink4a overexpression is associated with a profile associated with DNA damage, repair and senescence. Low levels of P16Ink4a induce reactive oxygen species (ROS) generation and increase endothelial cell leakage. Collectively, P16Ink4a represents an "antagonistic pleiotropy" gene, which is, on the one hand, required to prevent ROS generation and endothelial damage and, on the other hand, inhibits angiogenesis through induction of senescence at high levels.
Collapse
Affiliation(s)
| | | | - Nicole Wagner
- CNRS, INSERM, iBV, Université Côte d’Azur, 06107 Nice, France;
| |
Collapse
|
42
|
Supachawaroj N, Kerdmanee K, Limsitthichaikoon S. Lidocaine-Loaded Thermoresponsive Gel for Accelerated Wound Healing in Dry Socket and Oral Wounds. Gels 2024; 10:739. [PMID: 39590095 PMCID: PMC11594129 DOI: 10.3390/gels10110739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Dry socket, also known as alveolar osteitis, presents significant challenges in oral surgery because of severe pain and delayed wound healing. This study aims to address these challenges by developing and evaluating a lidocaine-loaded polyelectrolyte complex thermoresponsive gel (LG) designed to enhance wound healing and provide effective pain management in oral wounds. The thermoresponsive gel transitions from a liquid to a gel at body temperature, ensuring sustained contact with the wound site and prolonged release of lidocaine. The in vitro assessments, including cytotoxicity and wound scratch assays, demonstrated the biocompatibility and therapeutic potential of the LG formulation. Following this, palatal wounds were induced in rats, with healing monitored over a 14-days period. Histological analyses were conducted to assess tissue regeneration and inflammation. The results indicated that the LG formulation significantly improved wound closure rates, reduced inflammation, and accelerated epithelialization compared with control groups, primarily because of the high content of hyaluronic acid (HA). The synergistic effects of HA combined with the thermoresponsive properties of the gel facilitated faster healing. These findings suggest that LG is a promising therapeutic option for enhancing oral wound healing and effectively managing pain, particularly in conditions such as dry socket.
Collapse
Affiliation(s)
- Nuttawut Supachawaroj
- Department of Oral Surgery, College of Dental Medicine, Rangsit University, Pathum Thani 12000, Thailand;
| | - Kunchorn Kerdmanee
- Department of Periodontics, College of Dental Medicine, Rangsit University, Pathum Thani 12000, Thailand;
| | - Sucharat Limsitthichaikoon
- Department of Pharmaceutical Technology, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| |
Collapse
|
43
|
Hacımüftüoğlu E, Yesilyurt F, Babagil A, Mendil AS, Nadaroglu H. Rapid wound healing with silver nanoparticle-decorated miswak-derived carbon quantum dots. J Tissue Viability 2024; 33:968-977. [PMID: 39580240 DOI: 10.1016/j.jtv.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/13/2024] [Accepted: 10/28/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Oxidants are pivotal in combating aging, cancer, and weakened immunity. Shielding the body from free radicals is crucial for an extended lifespan. The development of effective systems for this purpose is paramount. This study investigated the impact of miswak carbon quantum dots (M-CQD) on radical elimination and wound healing. METHODS M-CQD were synthesized via a hydrothermal method using miswak water extract at 220 °C for 8 h. Chitosan-modified Ag@M-CS-CQD were produced. The characterization involved SEM, XRD, and FTIR analysis. M-CQD and Ag@M-CS-CQD emitted bluish-white light under UV excitation. RESULTS At 10, 30, and 50 μg/mL, DPPH• and phosphomolybdenum complex radicals inhibited 67 %, 82 %, and 98 %, respectively, with Ag@M-CS-CQD demonstrating the highest activity. The IC50 values for M-CQD and Ag@M-CS-CQD were 0.91 ± 0.010 and 1.183 ± 0.033, respectively. Ag@M-CS-CQD NPs displayed remarkable wound healing efficacy at 20 μg/L, highlighting their potential as potent antioxidants. Immunohistochemical analyzes also showed that Ag@M-CS-CQD increased TGF-β expression levels, which was associated with wound healing. CONCLUSION This eco-friendly synthesis method yielded Ag@M-CS-CQD, which exhibited robust antioxidant properties and efficacy in wound healing. This study emphasizes the potential of these nanoparticles as effective therapeutic agents.
Collapse
Affiliation(s)
- Esra Hacımüftüoğlu
- Atatürk University, Faculty of Theology, Basic Islamic Sciences, 25240, Erzurum, Turkey.
| | - Fatma Yesilyurt
- Ataturk University, Faculty of Medicine, Department of Medical Pharmacology, 25240, Erzurum, Turkey.
| | - Aynur Babagil
- Department of Nano-Science and Nano-Engineering, Institute of Science and Technology, Ataturk University, 25240, Erzurum, Turkey.
| | - Ali Sefa Mendil
- Department of Pathology, Faculty of Veterinary Medicine, Erciyes University, 38039, Kayseri, Turkey.
| | - Hayrunnisa Nadaroglu
- Department of Nano-Science and Nano-Engineering, Institute of Science and Technology, Ataturk University, 25240, Erzurum, Turkey; Department of Food Technology, Vocational College of Technical Science, Ataturk University, 25240, Erzurum, Turkey.
| |
Collapse
|
44
|
Hassan RM, Ali IH, El Kerdawy AM, Abo-Elfadl MT, Ghannam IAY. Novel benzenesulfonamides as dual VEGFR2/FGFR1 inhibitors targeting breast cancer: Design, synthesis, anticancer activity and in silico studies. Bioorg Chem 2024; 152:107728. [PMID: 39178704 DOI: 10.1016/j.bioorg.2024.107728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/04/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024]
Abstract
In the current study, a new series of benzenesulfonamides 6a-r was designed and synthesized as dual VEGFR-2 and FGFR1 kinase inhibitors with anti-cancer activity. The 4-trifluoromethyl benzenesulfonamide 6l exhibited the highest dual VEGFR-2/FGFR1 inhibitory activity with IC50 values of 0.025 and 0.026 µM, respectively. It showed a higher activity than sorafenib and staurosporine by 1.8- and 1.3-fold, respectively. Furthermore, compound 6l was further tested on EGFR and PDGFR-β kinases showing IC50 values of 0.106 and 0.077 µM, respectively. The target compounds were tested for their anticancer activity against NCI-60 panel of cancer cell lines at 10 µM concentration, where compound 6l displayed the highest mean growth inhibition percent % (GI%) of 60.38%. Compounds 6a, 6b, 6e, 6f, 6h-l, and 6n-r revealed promising GI% on breast cancer cell lines (MCF-7, T-47D, and MDA-MB-231), and were subjected to IC50 determination on these cell lines. The tested compounds showed a higher activity on T-47D and MCF-7 cell lines over MDA-MB-231 cell line compared to the used reference standard; sorafenib. Compounds 6e, 6h-j, 6l and 6o revealed IC50 values ≤ 20 µM against T-47D cell line, furthermore, they were found to be non-cytotoxic on Vero normal cell line. Furthermore, the effect of the most active compounds 6i, and 6l in T-47D cells on cell cycle analysis progression, cell apoptosis, and apoptosis markers was investigated. Both compounds arrested cell cycle progression at G1 phase, furthermore, they enhanced early and late apoptosis, as well as necrosis. The capability of compounds 6i, and 6l to induce apoptosis was further confirmed by their ability to raise BAX/BCl-2 ratio and caspase-3 level in the treated cells. Cell migration assay revealed that both compounds 6i and 6l have anti-migratory effects compared to control T-47D cells after 24, and 48 h. Molecular docking studies for compounds 6a-r on VEGFR-2 and FGFR1 binding sites showed that they exhibit an analogous binding mode in both target kinases which agrees with that of type II kinase inhibitors.
Collapse
Affiliation(s)
- Rasha M Hassan
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), P.O. 12622, Dokki, Giza, Egypt
| | - Islam H Ali
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Ahmed M El Kerdawy
- School of Pharmacy, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, United Kingdom; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Mahmoud T Abo-Elfadl
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo 12622, Egypt; Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki, Cairo, Egypt
| | - Iman A Y Ghannam
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt.
| |
Collapse
|
45
|
Kakizuka T, Natsume T, Nagai T. Compact lens-free imager using a thin-film transistor for long-term quantitative monitoring of stem cell culture and cardiomyocyte production. LAB ON A CHIP 2024. [PMID: 39436381 DOI: 10.1039/d4lc00528g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
With advancements in human induced pluripotent stem cell (hiPSC) technology, there is an increasing demand for quality control techniques to manage the long-term process of target cell production effectively. While monitoring systems designed for use within incubators are promising for assessing culture quality, existing systems still face challenges in terms of compactness, throughput, and available metrics. To address these limitations, we have developed a compact and high-throughput lens-free imaging device named INSPCTOR. The device is as small as a standard culture plate, which allows for the installation of multiple units within an incubator. INSPCTOR utilises a large thin-film transistor image sensor, enabling simultaneous observation of six independent culture environments, each approximately 1 cm2. With this device, we successfully monitored the confluency of hiPSC cultures and identified the onset timing of epithelial-to-mesenchymal transition during mesodermal induction. Additionally, we quantified the beating frequency and conduction of hiPSC-derived cardiomyocytes by using high-speed imaging modes. This enabled us to identify the onset of spontaneous beating during differentiation and assess chronotropic responses in drug evaluations. Moreover, by tracking beating frequency over 10 days of cardiomyocyte maturation, we identified week-scale and daily-scale fluctuations, the latter of which correlated with cellular metabolic activity. The metrics derived from this device would enhance the reproducibility and quality of target cell production.
Collapse
Affiliation(s)
- Taishi Kakizuka
- SANKEN, The University of Osaka, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan.
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, The University of Osaka, Yamadaoka 2-1, Suita, Osaka 565-0871, Japan
| | - Tohru Natsume
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, 2-3-26 Aoumi, Koto-ku, Tokyo 135-0064, Japan
| | - Takeharu Nagai
- SANKEN, The University of Osaka, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan.
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, The University of Osaka, Yamadaoka 2-1, Suita, Osaka 565-0871, Japan
| |
Collapse
|
46
|
Al Mousa AA, Abouelela ME, Mansour A, Nasr M, Ali YH, Al Ghamidi NS, Abo-Dahab Y, Mohamed H, Abo-Dahab NF, Hassane AMA. Wound Healing, Metabolite Profiling, and In Silico Studies of Aspergillus terreus. Curr Issues Mol Biol 2024; 46:11681-11699. [PMID: 39451574 PMCID: PMC11506626 DOI: 10.3390/cimb46100694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Burn injuries, which significantly affect global public health, require effective treatment strategies tailored to varying severity. Fungi are considered a sustainable, easily propagated source for lead therapeutic discovery. In this study, we explored the burn wound healing potential of Aspergillus terreus through a combination of in vitro, in vivo, metabolite profiling, and in silico analysis. The in vitro scratch assays performed with human skin fibroblast cells showed promising wound healing activity. Furthermore, the burn-induced rats model showed a marked improvement in cutaneous wound healing, evidenced by an accelerated rate of wound closure and better skin regeneration after A. terreus extract treatment at 14 days. The results of this study demonstrated significant enhancements in wound closure and tissue regeneration in the treated rat model, surpassing the outcomes of standard treatments. This controlled healing process, evidenced by superior collagen synthesis and angiogenesis and confirmed by histopathological studies, suggests that A. terreus has potential beyond the traditionally studied fungal metabolites. The metabolite profiling of 27 bioactive compounds was further investigated by docking analysis for the potential inhibition of the NF-κB pathway, which has an important function in inflammation and wound repair. The compounds eurobenzophenone A (7), aspernolide D (16), asperphenalenone A (23), aspergilate D (15), kodaistatin A (18), and versicolactone A (14) showed the highest binding affinity to the target protein with a pose score of -16.86, -14.65, -12.65, -12.45, -12.19, and -12.08 kcal/mol, respectively. Drug-likeness properties were also conducted. The findings suggest the potential wound healing properties of A. terreus as a source for lead therapeutic candidate discovery.
Collapse
Affiliation(s)
- Amal A. Al Mousa
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 145111, Riyadh 4545, Saudi Arabia;
| | - Mohamed E. Abouelela
- Department of Pharmacognosy, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo P.O. Box 11884, Egypt
| | - Ahmed Mansour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo P.O. Box 11884, Egypt;
| | - Mohamed Nasr
- Histology Department, Faculty of Medicine, Al-Azhar University, Cairo P.O. Box 11884, Egypt;
| | - Yasser H. Ali
- Department of Plastic & Reconstructive Surgery, Faculty of Medicine, Al-Azhar University, Cairo P.O. Box 11884, Egypt;
| | - Nadaa S. Al Ghamidi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 145111, Riyadh 4545, Saudi Arabia;
| | - Youssef Abo-Dahab
- Bioengineering and Therapeutic Sciences Department, University of California, P.O. Box 2520, San Francisco, CA 94158, USA;
| | - Hassan Mohamed
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut P.O. Box 71524, Egypt; (H.M.); (N.F.A.-D.); (A.M.A.H.)
- Colin Ratledge Center of Microbial Lipids, School of Agriculture Engineering and Food Science, Shandong University of Technology, Zibo 255000, China
| | - Nageh F. Abo-Dahab
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut P.O. Box 71524, Egypt; (H.M.); (N.F.A.-D.); (A.M.A.H.)
| | - Abdallah M. A. Hassane
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut P.O. Box 71524, Egypt; (H.M.); (N.F.A.-D.); (A.M.A.H.)
| |
Collapse
|
47
|
Awadelkareem AM, Patel M, Banu H, Adnan M. Integrating computational methods and i n vitro experimental validation reveals the pharmacological mechanism of Selaginella bryopteris (L.) Baker targeting major proteins in breast cancer. Heliyon 2024; 10:e38801. [PMID: 39430520 PMCID: PMC11489316 DOI: 10.1016/j.heliyon.2024.e38801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024] Open
Abstract
Breast cancer remains a significant global health challenge, necessitating the exploration of novel therapeutic options. The present study employs an integrated approach encompassing network pharmacology, molecular docking, molecular dynamics simulations, and in-vitro validation to investigate the potential of Selaginella bryopteris in breast cancer treatment. Initial network pharmacology analysis revealed different potential targets and pathways associated with breast cancer that could be modulated by S. bryopteris phytochemical constituents. Molecular docking and dynamics simulations further elucidated the stability and dynamics of protein-ligand complexes (lanaroflavone-EGFR and sequoiaflavone-CTNNB1). The in-vitro assays demonstrated the ability of S. bryopteris crude extract to inhibit cancer cell growth (IC50 - 78.34 μg/mL) migration and invasion, supporting the computational predictions. The integrated approach employed in the present study offers a robust framework for the systematic exploration of S. bryopteris in drug discovery as a promising candidate for breast cancer treatment.
Collapse
Affiliation(s)
- Amir Mahgoub Awadelkareem
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Mitesh Patel
- Research and Development Cell, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Humera Banu
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| |
Collapse
|
48
|
Gomes Daré R, Beatriz Chieco Costa A, Silva Martins T, Lopes LB. Simvastatin and adenosine-co-loaded nanostructured lipid carriers for wound healing: Development, characterization and cell-based investigation. Eur J Pharm Biopharm 2024:114533. [PMID: 39414092 DOI: 10.1016/j.ejpb.2024.114533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Chronic wounds represent a significant global health burden, characterized by delayed skin healing and associated comorbidities. The present study aimed to develop nanostructured lipid carriers (NLCs) as a topical delivery system for the co-administration of simvastatin and adenosine to address chronic wound management. The rationale behind the co-delivery approach was to mitigate the cytotoxicity associated with high-dose simvastatin, while preserving its therapeutic benefits through a potential synergistic or additive effect. A significant challenge in the development of these NLCs was the encapsulation of the highly hydrophilic adenosine within the hydrophobic lipid matrix. The NLCs were prepared using a hot homogenization-sonication method with a double emulsion technique and optimized through a series of formulation trials, employing various surfactants, solid and liquid lipids, to achieve efficient drug encapsulation, particularly for the hydrophilic adenosine. Optimized formulations F5- and F10-S/A 0.6 %/2 % (containing 0.6 % simvastatin and 2 % adenosine), exhibited promising physicochemical properties. The main difference was the liquid lipid used: F5 containing Miglyol 810 N, while F10 Capmul MCM C-8. Both formulations displayed a mean particle size below 230 nm, a polydispersity index (PDI) of approximately 0.2, and a zeta potential of around -22 mV. While simvastatin association efficiency (AE) was nearly 100 %, adenosine AE was higher for F10 (24 %), compared to F5 (13.5 %). F5 demonstrated superior stability compared to F10, maintaining consistent particle size and PDI over a 60-day period. Formulation F5 also demonstrated superior cell-based in vitro performance compared to F10, with higher cell viability (MTT assay), greater cell proliferation induction (SRB assay), and enhanced cell proliferation and migration in the wound-scratch assay. While F10 displayed higher adenosine AE, F5 excelled in terms of stability and biological activity. The slightly increase in intracellular reactive oxygen species levels observed with F5 may contribute to its enhanced proliferative effects. In-depth characterization revealed that F5 comprised spherical nanoparticles, and thermal analysis indicated no significant changes in the nanocarrier structure upon drug encapsulation. Additionally, ex vivo permeability study demonstrated superior skin retention of both simvastatin and adenosine for F5 compared to an emulsion control. Overall, the F5 nanocarrier demonstrated suitable physicochemical properties, cellular biocompatibility, induction of cell proliferation and migration events, and drug retention capacity in the skin layers, indicating its potential as a promising topical treatment for difficult-to-heal wounds.
Collapse
Affiliation(s)
- Regina Gomes Daré
- Institute of Biomedical Sciences, University of São Paulo, 1524 Professor Lineu Prestes Avenue, 05508-000 São Paulo, SP, Brazil.
| | - Ana Beatriz Chieco Costa
- Institute of Biomedical Sciences, University of São Paulo, 1524 Professor Lineu Prestes Avenue, 05508-000 São Paulo, SP, Brazil
| | - Tereza Silva Martins
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, 210 São Nicolau Street, 09913-030 Diadema, SP, Brazil
| | - Luciana B Lopes
- Institute of Biomedical Sciences, University of São Paulo, 1524 Professor Lineu Prestes Avenue, 05508-000 São Paulo, SP, Brazil.
| |
Collapse
|
49
|
Li Z, Li X, Seebacher NA, Liu X, Wu W, Yu S, Hornicek FJ, Huang C, Duan Z. CDK12 is a promising therapeutic target for the transcription cycle and DNA damage response in metastatic osteosarcoma. Carcinogenesis 2024; 45:786-798. [PMID: 39082894 DOI: 10.1093/carcin/bgae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 10/11/2024] Open
Abstract
Osteosarcoma (OS) is a bone malignant tumor affecting children, adolescents, and young adults. Currently, osteosarcoma is treated with chemotherapy regimens established over 40 years ago. The investigation of novel therapeutic strategies for the treatment of osteosarcoma remains an important clinical need. Cyclin-dependent kinases (CDKs) have been considered promising molecular targets in cancer therapy. Among these, CDK12 has been shown to play a crucial role in the pathogenesis of malignancies, but its clinical significance and biological mechanisms in osteosarcoma remain unclear. In the present study, we aim to determine the expression and function of CDK12 and evaluate its prognostic and therapeutic value in metastatic osteosarcoma. We found that overexpression of CDK12 was associated with high tumor grade, tumor progression and reduced patient survival. The underlying mechanism revealed that knockdown of CDK12 expression with small interfering RNA or functional inhibition with the CDK12-targeting agent THZ531 effectively exhibited time- and dose-dependent cytotoxicity. Downregulation of CDK12 paused transcription by reducing RNAP II phosphorylation, interfered with DNA damage repair with increased γH2AX, and decreased cell proliferation through the PI3K-AKT pathway. This was accompanied by the promotion of apoptosis, as evidenced by enhanced Bax expression and reduced Bcl-xL expression. Furthermore, the CDK12 selective inhibitor THZ531 also hindered ex vivo 3D spheroid formation, growth of in vitro 2D cell colony, and prevented cell mobility. Our findings highlight the clinical importance of CDK12 as a potentially valuable prognostic biomarker and therapeutic target in metastatic osteosarcoma.
Collapse
Affiliation(s)
- Zihao Li
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Xiaoyang Li
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Nicole A Seebacher
- Department of Oncology, University of Oxford OX3 9DU, Oxford, UK
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Xu Liu
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Wence Wu
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Shengji Yu
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Miami, FL, 33136USA
| | - Changzhi Huang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Miami, FL, 33136USA
| |
Collapse
|
50
|
Guha S, Talukdar D, Mandal GK, Mukherjee R, Ghosh S, Naskar R, Saha P, Murmu N, Das G. Crude extract of Ruellia tuberosa L. flower induces intracellular ROS, promotes DNA damage and apoptosis in triple negative breast cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118389. [PMID: 38821138 DOI: 10.1016/j.jep.2024.118389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ruellia tuberosa L. (Acanthaceae) is a weed plant traditionally used in folklore medicine as a diuretic, anti-hypertensive, anti-pyretic, anti-cancerous, anti-diabetic, analgesic, and gastroprotective agent. It has been previously reported that R. tuberosa L. is enriched with various flavonoids, exhibiting significant cytotoxic potential in various cancer models but a detailed study concerning its molecular mechanism is yet to be explored. AIM OF THE STUDY Exploring and validating R. tuberosa L. flower methanolic extract (RTME) as an anti-cancerous agent as per traditional usage with special emphasis on multi-drug resistant human triple-negative breast cancer (TNBC) and investigating the possible signaling networks and regulatory pathways involved in it. MATERIALS AND METHODS In this study, RTME was prepared using methanol, and phytochemical analysis was performed through GC-MS. Then, the extract was tested for its anti-cancer potential through in-vitro cytotoxicity assay, clonogenic assay, wound healing assay, ROS generation assay, cell cycle arrest, apoptotic nuclear morphology study, cellular apoptosis study, mitochondrial membrane potential (MMP) alteration study, protein, and gene expressions alteration study. In addition, toxicological status was evaluated in female Balb/C mice, and to check the receptor-ligand interactions, in-silico molecular docking was also conducted. RESULTS Several phytochemicals were found within RTME through GC-MS, which have been already reported to act as ROS inductive, DNA damaging, cell cycle arresting, and apoptotic agents against cancer cells. Moreover, RTME was found to exhibit significant in-vitro cytotoxicity along with a reduction in colony formation, and inhibition of cell migratory potential. It also induced intracellular ROS, promoted G0/G1 cell cycle arrest, caused mitochondrial membrane potential (MMP) alteration, and promoted cell death. The Western blot and qRT-PCR data revealed that RTME promoted the intrinsic pathway of apoptosis. Furthermore, blood parameters and organ histology on female Balb/C mice disclosed the non-toxic nature of RTME. Finally, an in-silico molecular docking study displayed that the three identified lead phytochemicals in RTME show strong receptor-ligand interactions with the anti-apoptotic Bcl-2 and give a clue to the possible molecular mechanism of the RTME extract. CONCLUSIONS RTME is a potential source of several phytochemicals that have promising therapeutic potential against TNBC cells, and thus could further be utilized for anti-cancer drug development.
Collapse
Affiliation(s)
- Subhabrata Guha
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India; Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Debojit Talukdar
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Gautam Kumar Mandal
- IQ City Medical College Hospital, IQ City Road, Durgapur, 713206, West Bengal, India.
| | - Rimi Mukherjee
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Srestha Ghosh
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Rahul Naskar
- Department of Chemistry, Jadavpur University, Kolkata, 700032, West Bengal, India.
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Nabendu Murmu
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Gaurav Das
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| |
Collapse
|