Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (57)

Search Parameters:
Keywords = anti-tubulin agents

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 1680 KiB  
Review
Microtubule-Targeting Agents: Advances in Tubulin Binding and Small Molecule Therapy for Gliomas and Neurodegenerative Diseases
by Maya Ezzo and Sandrine Etienne-Manneville
Int. J. Mol. Sci. 2025, 26(15), 7652; https://doi.org/10.3390/ijms26157652 (registering DOI) - 7 Aug 2025
Abstract
Microtubules play a key role in cell division and cell migration. Thus, microtubule-targeting agents (MTAs) are pivotal in cancer therapy due to their ability to disrupt cell division microtubule dynamics. Traditionally divided into stabilizers and destabilizers, MTAs are increasingly being repurposed for central [...] Read more.
Microtubules play a key role in cell division and cell migration. Thus, microtubule-targeting agents (MTAs) are pivotal in cancer therapy due to their ability to disrupt cell division microtubule dynamics. Traditionally divided into stabilizers and destabilizers, MTAs are increasingly being repurposed for central nervous system (CNS) applications, including brain malignancies such as gliomas and neurodegenerative diseases like Alzheimer’s and Parkinson’s. Microtubule-stabilizing agents, such as taxanes and epothilones, promote microtubule assembly and have shown efficacy in both tumour suppression and neuronal repair, though their CNS use is hindered by blood–brain barrier (BBB) permeability and neurotoxicity. Destabilizing agents, including colchicine-site and vinca domain binders, offer potent anticancer effects but pose greater risks for neuronal toxicity. This review highlights the mapping of nine distinct tubulin binding pockets—including classical (taxane, vinca, colchicine) and emerging (tumabulin, pironetin) sites—that offer new pharmacological entry points. We summarize the recent advances in structural biology and drug design, enabling MTAs to move beyond anti-mitotic roles, unlocking applications in both cancer and neurodegeneration for next-generation MTAs with enhanced specificity and BBB penetration. We further discuss the therapeutic potential of combination strategies, including MTAs with radiation, histone deacetylase (HDAC) inhibitors, or antibody–drug conjugates, that show synergistic effects in glioblastoma models. Furthermore, innovative delivery systems like nanoparticles and liposomes are enhancing CNS drug delivery. Overall, MTAs continue to evolve as multifunctional tools with expanding applications across oncology and neurology, with future therapies focusing on optimizing efficacy, reducing toxicity, and overcoming therapeutic resistance in brain-related diseases. Full article
(This article belongs to the Special Issue New Drugs Regulating Cytoskeletons in Human Health and Diseases)
Show Figures

Figure 1

17 pages, 7372 KiB  
Article
A Novel HDAC6 Inhibitor Enhances the Efficacy of Paclitaxel Against Ovarian Cancer Cells
by An-Jui Chi, Jui-Ling Hsu, Yun-Xin Xiao, Ji-Wang Chern, Jih-Hwa Guh, Chao-Wu Yu and Lih-Ching Hsu
Molecules 2025, 30(13), 2793; https://doi.org/10.3390/molecules30132793 - 28 Jun 2025
Viewed by 448
Abstract
Ovarian cancer cells overexpress HDAC6, and selective HDAC6 inhibitors have been considered potential new drugs for ovarian cancer either alone or in combination with other anticancer agents. We screened 46 potential novel HDAC6 inhibitors in ES-2 ovarian cancer cells and showed that compound [...] Read more.
Ovarian cancer cells overexpress HDAC6, and selective HDAC6 inhibitors have been considered potential new drugs for ovarian cancer either alone or in combination with other anticancer agents. We screened 46 potential novel HDAC6 inhibitors in ES-2 ovarian cancer cells and showed that compound 25253 demonstrated the most potent anti-proliferative activity and effective synergy with paclitaxel, which was also validated in TOV21G ovarian cancer cells. The combination of 25253 and paclitaxel significantly induced subG1 and apoptotic cells, revealed by PI staining assay and Annexin V-FITC/PI double staining assay, respectively. Western blot analysis showed downregulation of Bcl-2 and Bcl-XL, and upregulation of Bax and Bak, indicating that apoptosis was mediated through the intrinsic pathway. The combination increased γ-H2AX and p-p53 protein levels, suggesting the induction of DNA damage. Furthermore, HDAC6 was downregulated and acetylated α-tubulin was profoundly increased. Compound 25253 enhanced the inhibitory effect of paclitaxel on cell migration and invasion, possibly due to the extensive accumulation of acetylated α-tubulin, which affected microtubule dynamics. Taken together, the combination of 25253 and paclitaxel synergistically inhibited the growth, migration, and invasion of ovarian cancer cells and induced apoptosis, providing supporting evidence that the combination of HDAC6 inhibitors and paclitaxel may be a promising treatment strategy for ovarian cancer. Full article
(This article belongs to the Special Issue Innovative Anticancer Compounds and Therapeutic Strategies)
Show Figures

Graphical abstract

22 pages, 10936 KiB  
Article
TSPO Ligand 2-Cl-MGV-1 Mitigates Traumatic Brain Injury (TBI) in a Mouse Model
by Nasra Yasin, Leo Veenman, Beatriz Caballero, Nidal Zeineh, Laura Gonzalez-Blanco, Abraham Weizman and Moshe Gavish
Int. J. Mol. Sci. 2025, 26(10), 4854; https://doi.org/10.3390/ijms26104854 - 19 May 2025
Viewed by 1505
Abstract
In this study, we assessed the ability of 2-Cl-MGV-1 (2-chlorophenyl quinazolin-4-yl, dimethyl carbamate), a ligand of the 18 kDa mitochondrial translocator protein (TSPO), to mitigate brain damage in a mouse model of traumatic brain injury (TBI). TSPO is important for arresting the death [...] Read more.
In this study, we assessed the ability of 2-Cl-MGV-1 (2-chlorophenyl quinazolin-4-yl, dimethyl carbamate), a ligand of the 18 kDa mitochondrial translocator protein (TSPO), to mitigate brain damage in a mouse model of traumatic brain injury (TBI). TSPO is important for arresting the death of neurons and glia and counteracting microglial activation, and it provides anti-inflammatory activity, promotes regeneration (including neurons), and contributes to angiogenesis. We assessed the minimal dose of the TSPO ligand 2-Cl-MGV-1 that attenuates the magnitude of brain damage as well as the time window following TBI in which the treatment is effective. We found that 7.5 mg/kg of 2-Cl-MGV-1 can reduce the impact of the TBI as assessed by magnetic resonance imaging (MRI). We also found that 2-Cl-MGV-1 improved motor performance as observed in a treadmill test (80.9% fewer shocks needed and 40.7% more distance covered, both p < 0.05), and reduced anatomical brain damage (by 86.5%, p < 0.05), cell death (by 75.0%, p < 0.001), and microglial inflammatory response (by 50.2%, p < 0.01). The treatment also increased expression of neuronal markers NeuN and β3-tubulin (30.0%, p < 0.01; 36.0%, p < 0.01, respectively). The time window in which we found the treatment to be effective was 3–11 h after TBI. Our study suggests that agents active at the TSPO can significantly attenuate the outcome of TBI, including in the structural, cellular, and neuro-behavioral dimensions. The mechanisms involved in the attenuation of brain damage following TBI may be related to a decrease in cell death and to anti-inflammatory activity. TSPO seems to be a novel target for the development of agents aimed at the suppression of neurodegenerative processes. Full article
(This article belongs to the Special Issue Animal Research Model for Neurological Diseases, 2nd Edition)
Show Figures

Figure 1

18 pages, 2199 KiB  
Article
The Co-Administration of Paclitaxel with Novel Pyridine and Benzofuran Derivatives that Inhibit Tubulin Polymerisation: A Promising Anticancer Strategy
by Magdalena Perużyńska, Radosław Birger, Patrycja Kłos, Halina Kwiecień, Łukasz Struk, Jacek G. Sośnicki, Laurence Lafanechère and Marek Droździk
Pharmaceutics 2025, 17(2), 223; https://doi.org/10.3390/pharmaceutics17020223 - 9 Feb 2025
Viewed by 1145
Abstract
Background: Paclitaxel (PTX), a crucial microtubule-stabilising agent in cancer treatment, is limited by its adverse effects and hydrophobic nature, which necessitate the use of toxic solvents. This study proposes a novel approach combining PTX with new microtubule-destabilising compounds at low, safe doses [...] Read more.
Background: Paclitaxel (PTX), a crucial microtubule-stabilising agent in cancer treatment, is limited by its adverse effects and hydrophobic nature, which necessitate the use of toxic solvents. This study proposes a novel approach combining PTX with new microtubule-destabilising compounds at low, safe doses that are ineffective when used individually. Objective: The aim was to evaluate the therapeutic efficacy of combining PTX with previously described pyridine (S1, S22) and benzofuran derivatives (13b, 14), which have demonstrated promising anticancer properties by inhibiting microtubule polymerisation. Methods: The PrestoBlue assay was used to determine the optimal concentrations of each compound, enabling synergistic interactions with a low dose of PTX in HeLa cervical cancer cells. The combined effects of the compounds and PTX on apoptosis, cell cycle distribution, and mitotic spindle formation were then evaluated. Results: The results showed that compounds 13b (1 µM), 14 (0.1 µM), S1 (2 µM), and S22 (2 µM) enhanced the proapoptotic and antimitotic effects of 1 nM PTX, which was ineffective alone. Notably, live-cell imaging revealed that the concurrent use of S1 and PTX produced effects similar to those of a higher PTX concentration (5 nM). Conclusions: These findings suggest that these compounds enhance the anticancer efficacy of low-dose PTX, potentially paving the way for more effective and safer cancer therapies. Full article
(This article belongs to the Special Issue Combination Therapy Approaches for Cancer Treatment)
Show Figures

Graphical abstract

54 pages, 6031 KiB  
Article
(E)-1-(3-(3-Hydroxy-4-Methoxyphenyl)-1-(3,4,5-Trimethoxyphenyl)allyl)-1H-1,2,4-Triazole and Related Compounds: Their Synthesis and Biological Evaluation as Novel Antimitotic Agents Targeting Breast Cancer
by Gloria Ana, Azizah M. Malebari, Sara Noorani, Darren Fayne, Niamh M. O’Boyle, Daniela M. Zisterer, Elisangela Flavia Pimentel, Denise Coutinho Endringer and Mary J. Meegan
Pharmaceuticals 2025, 18(1), 118; https://doi.org/10.3390/ph18010118 - 17 Jan 2025
Cited by 2 | Viewed by 3879
Abstract
Background/Objectives: The synthesis of (E)-1-(1,3-diphenylallyl)-1H-1,2,4-triazoles and related compounds as anti-mitotic agents with activity in breast cancer was investigated. These compounds were designed as hybrids of the microtubule-targeting chalcones, indanones, and the aromatase inhibitor letrozole. Methods: A panel of [...] Read more.
Background/Objectives: The synthesis of (E)-1-(1,3-diphenylallyl)-1H-1,2,4-triazoles and related compounds as anti-mitotic agents with activity in breast cancer was investigated. These compounds were designed as hybrids of the microtubule-targeting chalcones, indanones, and the aromatase inhibitor letrozole. Methods: A panel of 29 compounds was synthesized and examined by a preliminary screening in estrogen receptor (ER) and progesterone receptor (PR)-positive MCF-7 breast cancer cells together with cell cycle analysis and tubulin polymerization inhibition. Results: (E)-5-(3-(1H-1,2,4-triazol-1-yl)-3-(3,4,5-trimethoxyphenyl)prop-1-en-1-yl)-2-methoxyphenol 22b was identified as a potent antiproliferative compound with an IC50 value of 0.39 mM in MCF-7 breast cancer cells, 0.77 mM in triple-negative MDA-MB-231 breast cancer cells, and 0.37 mM in leukemia HL-60 cells. In addition, compound 22b demonstrated potent activity in the sub-micromolar range against the NCI 60 cancer cell line panel including prostate, melanoma, colon, leukemia, and non-small cell lung cancers. G2/M phase cell cycle arrest and the induction of apoptosis in MCF-7 cells together with inhibition of tubulin polymerization were demonstrated. Immunofluorescence studies confirmed that compound 22b targeted tubulin in MCF-7 cells, while computational docking studies predicted binding conformations for 22b in the colchicine binding site of tubulin. Compound 22b also selectively inhibited aromatase. Conclusions: Based on the results obtained, these novel compounds are suitable candidates for further investigation as antiproliferative microtubule-targeting agents for breast cancer. Full article
Show Figures

Graphical abstract

15 pages, 4645 KiB  
Article
TBCC Domain-Containing Protein Regulates Sporulation and Virulence of Phytophthora capsici via Nutrient-Responsive Signaling
by Yaru Guo, Xiang Qiu, Bingting Lai, Caihuan Ou, Huirong Wang, Hengyuan Guo, Linying Li, Lili Lin, Dan Yu, Wenbo Liu and Justice Norvienyeku
Int. J. Mol. Sci. 2024, 25(22), 12301; https://doi.org/10.3390/ijms252212301 - 16 Nov 2024
Cited by 1 | Viewed by 1125
Abstract
Phytopathogenic oomycetes, particularly Phytophthora capsici, the causal agent of Phytophthora blight disease in essential vegetables and fruit crops, remains a persistent challenge in the vegetable production industry. However, the core molecular regulators of the pathophysiology and broad-range host characteristics of P. capsici [...] Read more.
Phytopathogenic oomycetes, particularly Phytophthora capsici, the causal agent of Phytophthora blight disease in essential vegetables and fruit crops, remains a persistent challenge in the vegetable production industry. However, the core molecular regulators of the pathophysiology and broad-range host characteristics of P. capsici remain unknown. Here, we used transcriptomics and CRISPR-Cas9 technology to functionally characterize the contributions of a novel gene (PcTBCC1) coding for a hypothetical protein with a tubulin-binding cofactor C domain with a putative chloroplast-targeting peptide (cTP) to the pathophysiological development of P. capsici. We observed significant upregulation in the expression of PcTBCC1 during pathogen–host interactions. However, the vegetative growth of the ∆Pctbcc1 strains was not significantly different from the wild-type strains. PcTBCC1 gene replacement significantly compromised the sporulation, pathogenic differentiation, and virulence of P. capsici. At the same time, ∆Pctbcc1 strains were sensitive to cell wall stress-inducing osmolytes. These observations, coupled with the close evolutionary ties between PcTBCC1 and pathogenic oomycetes and algae, partly support the notion that PcTBCC1 is a conserved determinant of pathogenesis. This study provides insights into the significance of tubulin-binding cofactors in P. capsici and underscores the potential of PcTbcc1 as a durable target for developing anti-oomycides to control phytopathogenic oomycetes. Full article
Show Figures

Figure 1

36 pages, 8405 KiB  
Article
Anti-Inflammatory and Cancer-Preventive Potential of Chamomile (Matricaria chamomilla L.): A Comprehensive In Silico and In Vitro Study
by Assia I. Drif, Rümeysa Yücer, Roxana Damiescu, Nadeen T. Ali, Tobias H. Abu Hagar, Bharati Avula, Ikhlas A. Khan and Thomas Efferth
Biomedicines 2024, 12(7), 1484; https://doi.org/10.3390/biomedicines12071484 - 5 Jul 2024
Cited by 3 | Viewed by 7030 | Correction
Abstract
Background and aim: Chamomile tea, renowned for its exquisite taste, has been appreciated for centuries not only for its flavor but also for its myriad health benefits. In this study, we investigated the preventive potential of chamomile (Matricaria chamomilla L.) towards cancer [...] Read more.
Background and aim: Chamomile tea, renowned for its exquisite taste, has been appreciated for centuries not only for its flavor but also for its myriad health benefits. In this study, we investigated the preventive potential of chamomile (Matricaria chamomilla L.) towards cancer by focusing on its anti-inflammatory activity. Methods and results: A virtual drug screening of 212 phytochemicals from chamomile revealed β-amyrin, β-eudesmol, β-sitosterol, apigenin, daucosterol, and myricetin as potent NF-κB inhibitors. The in silico results were verified through microscale thermophoresis, reporter cell line experiments, and flow cytometric determination of reactive oxygen species and mitochondrial membrane potential. An oncobiogram generated through comparison of 91 anticancer agents with known modes of action using the NCI tumor cell line panel revealed significant relationships of cytotoxic chamomile compounds, lupeol, and quercetin to microtubule inhibitors. This hypothesis was verified by confocal microscopy using α-tubulin-GFP-transfected U2OS cells and molecular docking of lupeol and quercetin to tubulins. Both compounds induced G2/M cell cycle arrest and necrosis rather than apoptosis. Interestingly, lupeol and quercetin were not involved in major mechanisms of resistance to established anticancer drugs (ABC transporters, TP53, or EGFR). Performing hierarchical cluster analyses of proteomic expression data of the NCI cell line panel identified two sets of 40 proteins determining sensitivity and resistance to lupeol and quercetin, further pointing to the multi-specific nature of chamomile compounds. Furthermore, lupeol, quercetin, and β-amyrin inhibited the mRNA expression of the proinflammatory cytokines IL-1β and IL6 in NF-κB reporter cells (HEK-Blue Null1). Moreover, Kaplan–Meier-based survival analyses with NF-κB as the target protein of these compounds were performed by mining the TCGA-based KM-Plotter repository with 7489 cancer patients. Renal clear cell carcinomas (grade 3, low mutational rate, low neoantigen load) were significantly associated with shorter survival of patients, indicating that these subgroups of tumors might benefit from NF-κB inhibition by chamomile compounds. Conclusion: This study revealed the potential of chamomile, positioning it as a promising preventive agent against inflammation and cancer. Further research and clinical studies are recommended. Full article
(This article belongs to the Special Issue Anticancer Activity and Metabolic Pathways of Natural Products 2.0)
Show Figures

Figure 1

13 pages, 3020 KiB  
Article
First-in-Class Humanized Antibody against Alternatively Spliced Tissue Factor Augments Anti-Metastatic Efficacy of Chemotherapy in a Preclinical Model of Pancreatic Ductal Adenocarcinoma
by Clayton S. Lewis, Charles Backman, Sabahat Ahsan, Ashley Cliff, Arthi Hariharan, Jen Jen Yeh, Xiang Zhang, Changchun Xie, Davendra P. S. Sohal and Vladimir Y. Bogdanov
Int. J. Mol. Sci. 2024, 25(5), 2580; https://doi.org/10.3390/ijms25052580 - 23 Feb 2024
Cited by 1 | Viewed by 1779
Abstract
Alternatively spliced tissue factor (asTF) promotes the progression of pancreatic ductal adenocarcinoma (PDAC) by activating β1-integrins on PDAC cell surfaces. hRabMab1, a first-in-class humanized inhibitory anti-asTF antibody we recently developed, can suppress PDAC primary tumor growth as a single agent. Whether hRabMab1 has [...] Read more.
Alternatively spliced tissue factor (asTF) promotes the progression of pancreatic ductal adenocarcinoma (PDAC) by activating β1-integrins on PDAC cell surfaces. hRabMab1, a first-in-class humanized inhibitory anti-asTF antibody we recently developed, can suppress PDAC primary tumor growth as a single agent. Whether hRabMab1 has the potential to suppress metastases in PDAC is unknown. Following in vivo screening of three asTF-proficient human PDAC cell lines, we chose to make use of KRAS G12V-mutant human PDAC cell line PaCa-44, which yields aggressive primary orthotopic tumors with spontaneous spread to PDAC-relevant anatomical sites, along with concomitant severe leukocytosis. The experimental design featured orthotopic tumors formed by luciferase labeled PaCa-44 cells; administration of hRabMab1 alone or in combination with gemcitabine/paclitaxel (gem/PTX); and the assessment of the treatment outcomes on the primary tumor tissue as well as systemic spread. When administered alone, hRabMab1 exhibited poor penetration of tumor tissue; however, hRabMab1 was abundant in tumor tissue when co-administered with gem/PTX, which resulted in a significant decrease in tumor cell proliferation; leukocyte infiltration; and neovascularization. Gem/PTX alone reduced primary tumor volume, but not metastatic spread; only the combination of hRabMab1 and gem/PTX significantly reduced metastatic spread. RNA-seq analysis of primary tumors showed that the addition of hRabMab1 to gem/PTX enhanced the downregulation of tubulin binding and microtubule motor activity. In the liver, hRabMab1 reduced liver metastasis as a single agent. Only the combination of hRabMab1 and gem/PTX eliminated tumor cell-induced leukocytosis. We here demonstrate for the first time that hRabMab1 may help suppress metastasis in PDAC. hRabMab1’s ability to improve the efficacy of chemotherapy is significant and warrants further investigation. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapies of Pancreatic Cancer)
Show Figures

Figure 1

12 pages, 2812 KiB  
Article
Tirbanibulin 1% Ointment for Actinic Keratosis: Results from a Real-Life Study
by Federica Li Pomi, Mario Vaccaro, Giovanni Pallio, Michelangelo Rottura, Natasha Irrera and Francesco Borgia
Medicina 2024, 60(2), 225; https://doi.org/10.3390/medicina60020225 - 28 Jan 2024
Cited by 12 | Viewed by 3122
Abstract
Background and Objectives: Tirbanibulin 1% ointment is a novel synthetic anti-proliferative agent that inhibits tubulin polymerization. It is approved for treating actinic keratosis (AK) on the face and scalp in adults. It has demonstrated good efficacy, an adequate safety profile and excellent [...] Read more.
Background and Objectives: Tirbanibulin 1% ointment is a novel synthetic anti-proliferative agent that inhibits tubulin polymerization. It is approved for treating actinic keratosis (AK) on the face and scalp in adults. It has demonstrated good efficacy, an adequate safety profile and excellent patient adherence in the phase 3 clinical trials, however data about its real-life efficacy and safety are lacking. Here we report the experience of the dermatology unit of the University Hospital of Messina. Materials and Methods: We performed a spontaneous open-label, prospective non-randomized study to assess the effectiveness and safety of tirbanibulin 1% ointment for the treatment of 228 AKs in 38 consecutive patients—28 males (73%) and 10 females (26%)—aged between 52 and 92 years (mean age: 72 ± 8.92 years). Results: Total clearance was recorded in 51% of lesions, while partial clearance was recorded in 73% of lesions. An excellent tolerability profile and high compliance rate were observed, with no treatment discontinuation due to the onset of adverse events. Conclusion: Our real-life experience confirms the effectiveness and safety of tirbanibulin ointment for the treatment of AKs. Full article
(This article belongs to the Section Dermatology)
Show Figures

Figure 1

28 pages, 6252 KiB  
Article
Frentizole, a Nontoxic Immunosuppressive Drug, and Its Analogs Display Antitumor Activity via Tubulin Inhibition
by Sergio Ramos, Alba Vicente-Blázquez, Marta López-Rubio, Laura Gallego-Yerga, Raquel Álvarez and Rafael Peláez
Int. J. Mol. Sci. 2023, 24(24), 17474; https://doi.org/10.3390/ijms242417474 - 14 Dec 2023
Cited by 2 | Viewed by 1904
Abstract
Antimitotic agents are one of the more successful types of anticancer drugs, but they suffer from toxicity and resistance. The application of approved drugs to new indications (i.e., drug repurposing) is a promising strategy for the development of new drugs. It relies on [...] Read more.
Antimitotic agents are one of the more successful types of anticancer drugs, but they suffer from toxicity and resistance. The application of approved drugs to new indications (i.e., drug repurposing) is a promising strategy for the development of new drugs. It relies on finding pattern similarities: drug effects to other drugs or conditions, similar toxicities, or structural similarity. Here, we recursively searched a database of approved drugs for structural similarity to several antimitotic agents binding to a specific site of tubulin, with the expectation of finding structures that could fit in it. These searches repeatedly retrieved frentizole, an approved nontoxic anti-inflammatory drug, thus indicating that it might behave as an antimitotic drug devoid of the undesired toxic effects. We also show that the usual repurposing approach to searching for targets of frentizole failed in most cases to find such a relationship. We synthesized frentizole and a series of analogs to assay them as antimitotic agents and found antiproliferative activity against HeLa tumor cells, inhibition of microtubule formation within cells, and arrest at the G2/M phases of the cell cycle, phenotypes that agree with binding to tubulin as the mechanism of action. The docking studies suggest binding at the colchicine site in different modes. These results support the repurposing of frentizole for cancer treatment, especially for glioblastoma. Full article
(This article belongs to the Special Issue Drug Repurposing: Emerging Approaches to Drug Discovery)
Show Figures

Figure 1

23 pages, 7293 KiB  
Article
Verubulin (Azixa) Analogues with Increased Saturation: Synthesis, SAR and Encapsulation in Biocompatible Nanocontainers Based on Ca2+ or Mg2+ Cross-Linked Alginate
by Kseniya N. Sedenkova, Denis N. Leschukov, Yuri K. Grishin, Nikolay A. Zefirov, Yulia A. Gracheva, Dmitry A. Skvortsov, Yanislav S. Hrytseniuk, Lilja A. Vasilyeva, Elena A. Spirkova, Pavel N. Shevtsov, Elena F. Shevtsova, Alina R. Lukmanova, Vasily V. Spiridonov, Alina A. Markova, Minh T. Nguyen, Alexander A. Shtil, Olga N. Zefirova, Alexander A. Yaroslavov, Elena R. Milaeva and Elena B. Averina
Pharmaceuticals 2023, 16(10), 1499; https://doi.org/10.3390/ph16101499 - 21 Oct 2023
Cited by 4 | Viewed by 2615
Abstract
Tubulin-targeting agents attract undiminished attention as promising compounds for the design of anti-cancer drugs. Verubulin is a potent tubulin polymerization inhibitor, binding to colchicine-binding sites. In the present work, a series of verubulin analogues containing a cyclohexane or cycloheptane ring 1,2-annulated with pyrimidine [...] Read more.
Tubulin-targeting agents attract undiminished attention as promising compounds for the design of anti-cancer drugs. Verubulin is a potent tubulin polymerization inhibitor, binding to colchicine-binding sites. In the present work, a series of verubulin analogues containing a cyclohexane or cycloheptane ring 1,2-annulated with pyrimidine moiety and various substituents in positions 2 and 4 of pyrimidine were obtained and their cytotoxicity towards cancer and non-cancerous cell lines was estimated. The investigated compounds revealed activity against various cancer cell lines with IC50 down to 1–4 nM. According to fluorescent microscopy data, compounds that showed cytotoxicity in the MTT test disrupt the normal cytoskeleton of the cell in a pattern similar to that for combretastatin A-4. The hit compound (N-(4-methoxyphenyl)-N,2-dimethyl-5,6,7,8-tetrahydroquinazolin-4-amine) was encapsulated in biocompatible nanocontainers based on Ca2+ or Mg2+ cross-linked alginate and it was demonstrated that its cytotoxic activity was preserved after encapsulation. Full article
Show Figures

Graphical abstract

25 pages, 10852 KiB  
Article
Exploring the Potential Biocontrol Isolates of Trichoderma asperellum for Management of Collar Rot Disease in Tomato
by C. Shanmugaraj, Deeba Kamil, Aditi Kundu, Praveen Kumar Singh, Amrita Das, Zakir Hussain, Robin Gogoi, P. R. Shashank, R. Gangaraj and M. Chaithra
Horticulturae 2023, 9(10), 1116; https://doi.org/10.3390/horticulturae9101116 - 10 Oct 2023
Cited by 8 | Viewed by 3226
Abstract
Bio-control agents are the best alternative to chemicals for the successful management of plant diseases. Among them, Trichoderma is commonly used as a biological control agent in plant disease management due to its ability to suppress soil-borne plant pathogens. In the present study, [...] Read more.
Bio-control agents are the best alternative to chemicals for the successful management of plant diseases. Among them, Trichoderma is commonly used as a biological control agent in plant disease management due to its ability to suppress soil-borne plant pathogens. In the present study, 20 Trichoderma asperellum isolates were collected from different geographical locations and confirmed using morphological characteristics and molecular phylogenetic inferences based on combined ITS and β-tubulin sequences. All twenty isolates were screened for their antagonism against the collar rot pathogen under in vitro and in planta conditions. The isolates were evaluated through dual culture and volatile methods in an in vitro study. Isolate A10 inhibited the test pathogen Agroathelia rolfsii at 94.66% in a dual culture assay and 70.95% in a volatile assay, followed by the isolates A11 and A17, which recorded 82.64% and 81.19% in dual culture assay and 63.75% and 68.27% in the volatile assay respectively. An in planta study was conducted under greenhouse conditions in tomato var. pusa ruby by pre- and post-inoculation of T. asperellum isolates in the A. rolfsii infected soil to evaluate their antagonistic potential against the disease. The A10 isolate was found effective under both pre- and post-inoculation conditions, with a disease inhibition percent of 86.17 and 80.60, respectively, followed by the isolates A11 and A17, which exhibited inhibition of 77.80% and 75.00% in pre-inoculation and 72.22% and 69.44% in post-inoculation, respectively. Further, biochemical analysis was conducted to determine the specific activity of hydrolytic enzymes produced by T. asperellum during interaction with A. rolfsii. We found that isolate A10 produces more hydrolytic enzymes with the specific activity of 174.68 IU/mg of β-1,3 glucanase, 183.48 IU/mg of β-1, 4 glucanase, 106.06 IU/mg of protease, followed by isolate A17, A11 respectively. In GC-MS analysis, we observed maximum anti-microbial volatile organic compounds from the isolate A10, including 2H-Pyran-2-one (17.39%), which was found to be most abundant, followed by dienolactone (8.43%), α-pyrone (2.19%), and harziandione (0.24%) respective retention time of 33.48, 33.85, 33.39, and 64.23 min, respectively, compared to other isolates. In the TLC assay, we observed that a greater number of bands were produced by the A10 and A17 isolates in the Hexane: Ethyl Acetate (1:1) solvent system than in the 9:1 solvent system, which represents the presence of major metabolites in the ethyl acetate extract. Full article
Show Figures

Figure 1

11 pages, 1984 KiB  
Article
Avermectin B1a Shows Potential Anti-Proliferative and Anticancer Effects in HCT-116 Cells via Enhancing the Stability of Microtubules
by Qendresa Hoti, Duygu Gencalp Rustem and Ozlem Dalmizrak
Curr. Issues Mol. Biol. 2023, 45(8), 6272-6282; https://doi.org/10.3390/cimb45080395 - 27 Jul 2023
Cited by 5 | Viewed by 2910
Abstract
Avermectins are a group of macrocyclic lactones that are commonly used as pesticides to treat pests and parasitic worms. Some members of the avermectin family, such as ivermectin, have been found to exhibit anti-proliferative activity toward cancer cells. This study aimed to investigate [...] Read more.
Avermectins are a group of macrocyclic lactones that are commonly used as pesticides to treat pests and parasitic worms. Some members of the avermectin family, such as ivermectin, have been found to exhibit anti-proliferative activity toward cancer cells. This study aimed to investigate the potential anti-cancer activities of avermectin B1a using the HCT-116 colon cancer cell line. The MTT assay was used to calculate the IC50 by incubating cells with increasing doses of avermectin B1a for 24, 48, and 72 h. Flow cytometry was used to evaluate apoptosis following the 24 h incubation of cells. The migration capacity of the HCT-116 cells in the absence or presence of avermectin B1a was also investigated. Finally, tubulin polymerization in the presence of avermectin B1a was evaluated. Avermectin B1a presented anti-proliferative activity with an IC50 value of 30 μM. Avermectin B1a was found to promote tubulin polymerization at 30 μM. In addition, avermectin B1a induced apoptosis in HCT-116 cells and substantially diminished their ability to migrate. Avermectin B1a exhibits significant anti-cancer activity and enhances tubulin polymerization, suggesting that it can be used as a promising microtubule-targeting agent for the development of future anticancer drugs. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

29 pages, 4751 KiB  
Article
Novel Tetrazole Derivatives Targeting Tubulin Endowed with Antiproliferative Activity against Glioblastoma Cells
by Laura Gallego-Yerga, Andrea Jazmín Chiliquinga and Rafael Peláez
Int. J. Mol. Sci. 2023, 24(13), 11093; https://doi.org/10.3390/ijms241311093 - 4 Jul 2023
Cited by 4 | Viewed by 2645
Abstract
Increasing awareness of the structure of microtubules has made tubulin a relevant target for the research of novel chemotherapies. Furthermore, the particularly high sensitivity of glioblastoma multiforme (GBM) cells to microtubule disruption could open new doors in the search for new anti-GBM treatments. [...] Read more.
Increasing awareness of the structure of microtubules has made tubulin a relevant target for the research of novel chemotherapies. Furthermore, the particularly high sensitivity of glioblastoma multiforme (GBM) cells to microtubule disruption could open new doors in the search for new anti-GBM treatments. However, the difficulties in developing potent anti-tubulin drugs endowed with improved pharmacokinetic properties necessitates the expansion of medicinal chemistry campaigns. The application of an ensemble pharmacophore screening methodology helped to optimize this process, leading to the development of a new tetrazole-based tubulin inhibitor. Considering this scaffold, we have synthesized a new family of tetrazole derivatives that achieved remarkable antimitotic effects against a broad panel of cancer cells, especially against GBM cells, showing high selectivity in comparison with non-tumor cells. The compounds also exerted high aqueous solubility and were demonstrated to not be substrates of efflux pumps, thus overcoming the main limitations that are usually associated with tubulin binding agents. Tubulin polymerization assays, immunofluorescence experiments, and flow cytometry studies demonstrated that the compounds target tubulin and arrest cells at the G2/M phase followed by induction of apoptosis. The docking experiments agreed with the proposed interactions at the colchicine site and explained the structure–activity relationships. Full article
(This article belongs to the Special Issue Advances in Drug Discovery and Synthesis)
Show Figures

Figure 1

31 pages, 3979 KiB  
Article
Phenothiazine- and Carbazole-Cyanochalcones as Dual Inhibitors of Tubulin Polymerization and Human Farnesyltransferase
by Andreea Zubaș, Alina Ghinet, Amaury Farce, Joëlle Dubois and Elena Bîcu
Pharmaceuticals 2023, 16(6), 888; https://doi.org/10.3390/ph16060888 - 16 Jun 2023
Cited by 3 | Viewed by 2327
Abstract
In the search for innovative approaches to cancer chemotherapy, a chemical library of 49 cyanochalcones, 1a-r, 2a-o, and 3a-p, was designed as dual inhibitors of human farnesyltransferase (FTIs) and tubulin polymerization (MTIs) (FTIs/MTIs), two important biological targets in oncology. This [...] Read more.
In the search for innovative approaches to cancer chemotherapy, a chemical library of 49 cyanochalcones, 1a-r, 2a-o, and 3a-p, was designed as dual inhibitors of human farnesyltransferase (FTIs) and tubulin polymerization (MTIs) (FTIs/MTIs), two important biological targets in oncology. This approach is innovative since the same molecule would be able to interfere with two different mitotic events of the cancer cells and prevent these cells from developing an emergency route and becoming resistant to anticancer agents. Compounds were synthesized by the Claisen–Schmidt condensation of aldehydes with N-3-oxo-propanenitriles under classical magnetic stirring and under sonication. Newly synthesized compounds were screened for their potential to inhibit human farnesyltransferase, tubulin polymerization, and cancer cell growth in vitro. This study allowed for the identification of 22 FTIs and 8 dual FTIs/MTIs inhibitors. The most effective molecule was carbazole-cyanochalcone 3a, bearing a 4-dimethylaminophenyl group (IC50 (h-FTase) = 0.12 µM; IC50 (tubulin) = 0.24 µM) with better antitubulin activity than the known inhibitors that were previously reported, phenstatin and (-)-desoxypodophyllotoxin. The docking of the dual inhibitors was realized in both the active site of FTase and in the colchicine binding site of tubulin. Such compounds with a dual inhibitory profile are excellent clinical candidates for the treatment of human cancers and offer new research perspectives in the search for new anti-cancer drugs. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

Back to TopTop