1
|
Bejcek LP, Eli OS, Kapkayeva DM, Nafie J, Beutler JA, Gallicchio E, Sackett DL, Murelli RP. Deconstruction of Desacetamidocolchicine's B Ring Reveals a Class 3 Atropisomeric AC Ring with Tubulin Binding Properties. J Org Chem 2025. [PMID: 40422813 DOI: 10.1021/acs.joc.5c00284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Colchicine is one of the oldest known microtubule-targeting agents and also represents a classic example of axial chirality and atropisomerism in medicine. This is because colchicine's axially chiral methoxytropone-trimethoxybenzene (called the AC ring) is directly responsible for tubulin binding and is thermodynamically set into the requisite aR form by a point chiral acetamido group on its B ring. Indeed, desacetamidocolchicine (DAAC), a colchicine analogue without the acetamido group, racemizes within minutes. Herein, we describe the synthesis as well as physical and biological characterization of a series of AC ring-containing molecules that represent B-ring further deconstructed variants of DAAC. These studies revealed a novel analogue with an AC ring that is highly stable to epimerization based not on thermodynamic stabilization but rather a high rotational barrier energy. Profiling and characterization of the dihedral angles were carried out computationally and experimentally using vibrational circular dichroism, demonstrating that the ground state dihedral angles of the new molecules differ significantly from those of colchicine. However, despite this difference, the molecule retained antiproliferative, tubulin-binding, and tubulin polymerization inhibitory activity.
Collapse
Affiliation(s)
- Lauren P Bejcek
- PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Department of Chemistry and Biochemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Orugbani S Eli
- PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Department of Chemistry and Biochemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Diana M Kapkayeva
- PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| | - Jordan Nafie
- Biotools, Inc., 17546 Bee Line Highway, Jupiter, Florida 33478, United States
| | - John A Beutler
- Molecular Targets Program, National Cancer Institute, National Institutes of Health. 1050 Boyles Street, Frederick, Maryland 21702, United States
| | - Emilio Gallicchio
- PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Department of Chemistry and Biochemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- The Graduate Center of the City University of New York, PhD Program in Biochemistry, New York, New York 10016, United States
| | - Dan L Sackett
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 1 Center Dr, Bethesda, Maryland 20892, United States
| | - Ryan P Murelli
- PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Department of Chemistry and Biochemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- The Graduate Center of the City University of New York, PhD Program in Biochemistry, New York, New York 10016, United States
| |
Collapse
|
2
|
Beccari MS, Arnold-Garcia O, Baughn MW, Artates JW, McAlonis-Downes M, Lim J, Leyva-Cázares DF, Rubio-Lara HI, Ramirez-Rodriguez A, Bernal-Buenrostro CN, Murgia-Bay B, Rangel CK, Kim DH, Melamed Z, Lutz CM, Lagier-Tourenne C, Corbett KD, López-Erauskin J, Cleveland DW. Stathmin-2 enhances motor axon regeneration after injury independent of its binding to tubulin. Proc Natl Acad Sci U S A 2025; 122:e2502294122. [PMID: 40392845 DOI: 10.1073/pnas.2502294122] [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: 02/09/2025] [Accepted: 04/19/2025] [Indexed: 05/22/2025] Open
Abstract
Stathmin-2 (also known as SCG10) is encoded by the STMN2 gene, whose mRNA is one of the most abundantly expressed in human motor neurons. In almost all instances of ALS and other TDP-43 proteinopathies, stathmin-2 encoding mRNAs are cryptically spliced and polyadenylated in motor neurons, a pathogenic consequence of nuclear loss of function of the RNA binding protein TDP-43. While stathmin-2 has been shown to enhance regeneration after axonal injury to axons of cultured motor neurons, here, we show that after crush injury within the adult murine nervous system of wild-type or stathmin-2-null mice, the presence of stathmin-2 reduces axonal and neuromuscular junction degeneration and stimulates reinnervation and functional recovery. Mechanistically, although stathmin-2 has been proposed to function through direct binding to α/β tubulin heterodimers and correspondingly to affect microtubule assembly and dynamics, stathmin-2's role in axon regeneration after axotomy is shown to be independent of its tubulin binding abilities.
Collapse
Affiliation(s)
- Melinda S Beccari
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Olatz Arnold-Garcia
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Neurosciences, Biogipuzkoa Health Research Institute, San Sebastián 20014, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Carlos III Institute (ISCIII), Spanish Ministry of Sciences and Innovation, Madrid 28029, Spain
| | - Michael W Baughn
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Jonathan W Artates
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Melissa McAlonis-Downes
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Jaisen Lim
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Dulce Fernanda Leyva-Cázares
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
- Programa de Licenciatura en Medicina, Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Baja California 22427, México
| | - Hugo Isaac Rubio-Lara
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
- Programa de Licenciatura en Medicina, Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Baja California 22427, México
| | - Andrea Ramirez-Rodriguez
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
- Programa de Licenciatura en Medicina, Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Baja California 22427, México
| | - Carol N Bernal-Buenrostro
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
- Programa de Licenciatura en Medicina, Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Baja California 22427, México
| | - Brian Murgia-Bay
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
- Programa de Licenciatura en Medicina, Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Baja California 22427, México
| | - Carolina K Rangel
- Prinses Maxima Centrum voor Kinderoncologie, Utrecht 3584 CS, The Netherlands
| | - Dong Hyun Kim
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Ze'ev Melamed
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Cathleen M Lutz
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME 04609
| | - Clotilde Lagier-Tourenne
- Department of Neurology, Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Broad Institute of Harvard University and MIT, Cambridge, MA 02142
| | - Kevin D Corbett
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093
| | - Jone López-Erauskin
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Don W Cleveland
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
3
|
Cohen A, Ben-Nun I, Dharan R, Tayri-Wilk T, Shemesh A, Ginsburg A, Millgram A, Levi-Kalisman Y, Ringel I, Raviv U. Modulating the Curvature of Protein Self-Assembled Spiral Nanotubules. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29146-29157. [PMID: 40354519 PMCID: PMC12100598 DOI: 10.1021/acsami.5c01405] [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] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/15/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
Structural transformations from ribbons to twisted ribbons to helical ribbons are often observed across supramolecular assemblies and macroscopic structures and can be described under a consistent theoretical framework. Conical molecular self-assembled structures, however, are rarely observed, may require more than one subunit, their dimensions are hard to control, and are poorly understood. Cytoskeleton microtubule (MT) is a dynamic protein-polymer that self-assembles from αβ-tubulin heterodimer, providing mechanical support to Eukaryotic cells. Colchicine is a drug known to bind the exchangeable nucleotide site on the β-tubulin subunit and suppress MT assembly. The tetravalent polyamine spermine promotes MT assembly and tubulin spiral structures, including conical tubulin spirals, tubules of conical spirals, and inverted helical tubules. Here we show how colchicine as a single agent suppressed MT and tubulin single ring assembly already at substoichiometric concentrations, whereas in the presence of spermine, the tubulin-colchicine stoichiometry controlled the dimensions and curvature of tubulin spiral assemblies. At a fixed spermine concentration, the concentration of colchicine modulated the radii of the nanotubular structures. The radii of the inverted helical nanotubules and conical spiral nanotubules monotonically decreased with colchicine concentration. We attribute our observation to the increased curvature of the tubulin dimer subunit induced by colchicine.
Collapse
Affiliation(s)
- Ariel Cohen
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
| | - Itai Ben-Nun
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
| | - Raviv Dharan
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
| | - Tamar Tayri-Wilk
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
| | - Asaf Shemesh
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
| | - Avi Ginsburg
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
| | - Abigail Millgram
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
| | - Yael Levi-Kalisman
- The
Harvey M. Krueger Family Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
| | - Israel Ringel
- Institute
for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Karem, Jerusalem9112102, Israel
| | - Uri Raviv
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
- The
Harvey M. Krueger Family Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem9190401, Israel
| |
Collapse
|
4
|
Ahsan MJ, Kumar V, Ali A, Ali A, Yusuf M, Ahmad I, Patel H, Salahuddin, Ahsan MF. Design and synthesis of newer 5-aryl- N-(naphthalen-2-yl)-1,3,4-oxadiazol-2-amine analogues as anticancer agents. Future Med Chem 2025:1-12. [PMID: 40371593 DOI: 10.1080/17568919.2025.2504335] [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: 03/08/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
AIM Cancer is the second leading cause of death and chemotherapy is widely used and well-known for treating cancer, yet it has lots of adverse side effects, making the search for novel compounds imperative. We reported here design, synthesis, DFT analysis, anticancer evaluation and in-silico studies of new 1,3,4-oxadiazoles (4a-e). MATERIAL AND METHODS IMC-038525 and IMC-094332 tubulin inhibitors' oxadiazole-linked aryl cores inspired the innovative compounds, and synthesis was accomplished in two steps followed by their characterization by spectral data. The HOMO and LUMO energy gap (ΔE) was determined to investigate compounds' (4a-e) stability followed by their anticancer activity at 10 μM and in-silico studies. RESULTS AND CONCLUSION 5-(4-Nitrophenyl)-N-(naphthalene-2-yl)-1,3,4-oxadiazol-2-amine (4b) demonstrated substantial anticancer activity against a few cell lines like SR, MDA-MB-435, MOLT-4, K-562, and HL-60(TB). 5-(3,4,5-Trimethoxyphenyl)-N-(naphthalene-2-yl)-1,3,4-oxadiazol-2-amine (4e) demonstrated promising anticancer activity against cell lines, UO-31, NCI-H226, CAKI-1, PC-3, and MCF7. The molecular docking against tubulin's colchicine binding site (PDB ID: 1AS0), displayed a docking score of -7.295 Kcal/mol and a H-bond interaction with Ala317 residue for the ligand 4e. The ligand 4e was found to interacted 24 amino acids of the tubulin protein in MD simulation investigation with moderate local conformational changes with ligand 4e (< 1 Å).
Collapse
Affiliation(s)
- Mohamed Jawed Ahsan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jahangirabad Institute of Technology, Barabanki, Uttar Pradesh, India
| | - Vivek Kumar
- Department of Pharmaceutical Chemistry, Maharishi Arvind College of Pharmacy, Ambabari Circle, Jaipur, Rajasthan, India
| | - Amena Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Abuzer Ali
- Department of Pharmacognosy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Mohammad Yusuf
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Salahuddin
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering & Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Md Faiyaz Ahsan
- Department of Chemistry, Bihar National College, Patna, Bihar, India
| |
Collapse
|
5
|
Chagas C, da Silva EB, Alves BDCA, da Veiga GL, Pereira EC, Haddad P, Schumacher ML, Britos TN, Lima LM, Barreiro EJL, Ferreira FF, Fonseca FLA. Synthesis, Characterization, and Toxicity Assessment of Superparamagnetic Iron Oxide Nanoparticles Coated with Antitumor Molecules. ACS OMEGA 2025; 10:17237-17248. [PMID: 40352520 PMCID: PMC12060056 DOI: 10.1021/acsomega.4c09185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/26/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
New definitions describe cancer as a condition where cells, transformed by natural selection, undergo uncontrolled proliferation. Currently, several available drugs for treating cancer present side effects and are nonselective, thus complicating patient treatment. To overcome such issues, alternative therapies using inorganic nanoparticles and parent compounds are becoming more attractive. Here, we demonstrate the use of biocompatible superparamagnetic iron oxide nanoparticles (SPIONs) coated with the LASSBio-1735 parent compound in BALB/c mice inoculated with the Ehrlich tumor. We first characterize the formed nanoparticles and confirm and quantify the anchoring of LASSBio-1735 on the surface of the SPIONs. Tests conducted after 7, 14, and 30 days show that no significant alterations were found in erythrocyte count analysis, hemoglobin and hematocrit determination, platelet and leukocyte counts, neutrophil/lymphocyte ratio, serum iron dosage, alanine aminotransferase, aspartate aminotransferase, serum creatinine, and urea determination, as well as analysis of the animals' and tumor weights. This confirms that this new alternative can be considered in clinical trials to treat solid tumors.
Collapse
Affiliation(s)
- Camila Chagas
- Clinical
Analysis Laboratory of the Centro Universitário FMABC, Av. Príncipe de Gales, 821,
Bairro Vila Príncipe de Gales, Santo
André, São Paulo 09060-650, Brazil
| | - Emerson B. da Silva
- Clinical
Analysis Laboratory of the Centro Universitário FMABC, Av. Príncipe de Gales, 821,
Bairro Vila Príncipe de Gales, Santo
André, São Paulo 09060-650, Brazil
| | - Beatriz da C. A. Alves
- Clinical
Analysis Laboratory of the Centro Universitário FMABC, Av. Príncipe de Gales, 821,
Bairro Vila Príncipe de Gales, Santo
André, São Paulo 09060-650, Brazil
| | - Glaucia L. da Veiga
- Clinical
Analysis Laboratory of the Centro Universitário FMABC, Av. Príncipe de Gales, 821,
Bairro Vila Príncipe de Gales, Santo
André, São Paulo 09060-650, Brazil
| | - Edimar C. Pereira
- Chemistry
Department, Federal University of São
Paulo—Campus Diadema, Rua São Nicolau, 210, Centro, Diadema, São Paulo 09913-030, Brazil
| | - Paula Haddad
- Chemistry
Department, Federal University of São
Paulo—Campus Diadema, Rua São Nicolau, 210, Centro, Diadema, São Paulo 09913-030, Brazil
| | - Maria Lúcia Schumacher
- Chemistry
Department, Federal University of São
Paulo—Campus Diadema, Rua São Nicolau, 210, Centro, Diadema, São Paulo 09913-030, Brazil
| | - Tatiane Nassar Britos
- Chemistry
Department, Federal University of São
Paulo—Campus Diadema, Rua São Nicolau, 210, Centro, Diadema, São Paulo 09913-030, Brazil
| | - Lídia M. Lima
- LASSBio,
Institute of Biomedical Sciences, Federal University of Rio de Janeiro
(UFRJ), Sala 35—Prédio Do Centro de Ciências
da Saúde, Av.
Carlos Chagas, 373—Bloco K, 2° Andar, Cidade Universitária,
Ilha do Fundão, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Graduate
Program of Chemistry, Institute of Chemistry, Federal University of
Rio de Janeiro (UFRJ), Av. Athos da Silveira Ramos, no 149, Bloco A—7° Andar,
Centro de Tecnologia, Cidade Universitária, Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Eliezer J. L. Barreiro
- LASSBio,
Institute of Biomedical Sciences, Federal University of Rio de Janeiro
(UFRJ), Sala 35—Prédio Do Centro de Ciências
da Saúde, Av.
Carlos Chagas, 373—Bloco K, 2° Andar, Cidade Universitária,
Ilha do Fundão, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Fabio F. Ferreira
- Center
for Natural and Human Sciences (CCNH), Federal
University of ABC (UFABC), Santo
André, São Paulo 09280-560, Brazil
- Nanomedicine
Research Unit (NANOMED), Federal University
of ABC (UFABC), Santo André, São Paulo 09280-560, Brazil
| | - Fernando L. A. Fonseca
- Clinical
Analysis Laboratory of the Centro Universitário FMABC, Av. Príncipe de Gales, 821,
Bairro Vila Príncipe de Gales, Santo
André, São Paulo 09060-650, Brazil
- Chemistry
Department, Federal University of São
Paulo—Campus Diadema, Rua São Nicolau, 210, Centro, Diadema, São Paulo 09913-030, Brazil
| |
Collapse
|
6
|
Shaikh S, Hamza M, Neppala S, Singh S, Upreti P, Umer AM, Manish KC, Pandya K, Bahar Y, Sattar Y, Alraies MC. Colchicine for secondary prevention in patients with acute coronary syndrome: A systematic review and meta-analysis. Int J Cardiol 2025; 425:133045. [PMID: 39923944 DOI: 10.1016/j.ijcard.2025.133045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/12/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Despite optimal therapy, coronary artery disease (CAD) remains a significant public health concern worldwide. Studies have increasingly recognized the role of inflammation in atherosclerosis. Colchicine, a potent anti-inflammatory drug commonly used to treat gout, and pericarditis is being evaluated in this study for its safety and efficacy in preventing CAD following an acute coronary syndrome (ACS). METHODS We searched PubMed and Embase for studies up to April 2024 comparing colchicine to standard medical treatment in ACS patients. Primary outcomes included major adverse cardiovascular events (MACE) and recurrent ACS, while secondary outcomes were cardiovascular death, congestive heart failure (CHF), stroke, hospitalizations, and gastrointestinal (GI) side effects. Data were pooled using a random-effects model. RESULTS We included nine studies with a pooled sample size of 7260 patients. The mean age was 60.1 (±11.8) years, with 19.3 % females and a mean follow-up duration of 8.5 (±6) months. Patients who received colchicine treatment demonstrated a reduced risk of re-hospitalizations (OR 0.52 [0.34-0.81]) but had increased GI effects (OR 2.10 [1.20-3.68]). There was no significant difference in cardiovascular death (OR 1.17 [0.52-2.63]), MACE (OR 0.68 [0.45-1.01]), stroke (OR 0.46 [0.18-1.18]), recurrent ACS (OR 0.55 [0.28-1.09]) and the incidence of CHF (OR 0.90 [0.38-2.12]) between patients treated with colchicine versus standard medical treatment. CONCLUSION Adding colchicine to standard medical therapy in ACS patients significantly reduced hospitalizations but is associated with increased GI side effects. Further prospective trials are required to validate these findings and determine if early intervention with colchicine treatment improves clinical outcomes in ACS patients.
Collapse
Affiliation(s)
- Safia Shaikh
- Washington University in St Louis, St Louis, MO, USA
| | | | | | | | - Prakash Upreti
- Sands-Constellation Heart Institute, Rochester Regional Health, Rochester, NY, USA
| | | | - K C Manish
- North Alabama Medical Center, Florence, AL, USA
| | | | | | | | - M Chadi Alraies
- Cardiovascular Institute, Detroit Medical Center, Detroit, MI, USA.
| |
Collapse
|
7
|
Ashimbayeva M, Szakonyi Z, Adekenov SM, Szemerédi N, Spengler G, Le TM. Synthesis and Antiproliferative Effects of Grossheimin-Derived Aminoanalogues. Biomolecules 2025; 15:578. [PMID: 40305367 PMCID: PMC12024577 DOI: 10.3390/biom15040578] [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: 02/11/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025] Open
Abstract
Grossheimin, a guaiane-type sesquiterpene lactone, displayed a diverse range of biological activities, including anticancer, anti-inflammatory and antimicrobial effects. Various amino analogues of grossheimin were prepared through a Michael addition at its highly active α-methylene-γ-lactone motif. On the other hand, grossheimin was reduced to diol, which was then subjected to nucleophilic addition or acetylation to introduce heteroatoms associated with oxygen, sulfur or nitrogen functionalities. All of the synthesised Michael and acetylated adducts were evaluated for their in vitro cytotoxic action on human colon adenocarcinoma lines, including Colo205 and Colo320. The bioassay results indicated that the acetylated adducts displayed a potent cytotoxic effect compared to grossheimin, the parent molecule. A docking study was also performed to exploit the observed results.
Collapse
Affiliation(s)
- Meruyert Ashimbayeva
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
| | - Zsolt Szakonyi
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
| | - Sergazy M. Adekenov
- JSC Research and Production Center “Phytochemistry”, Karaganda 100009, Kazakhstan;
| | - Nikoletta Szemerédi
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary; (N.S.); (G.S.)
| | - Gabriella Spengler
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary; (N.S.); (G.S.)
| | - Tam Minh Le
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
- HUN-REN–SZTE Stereochemistry Research Group, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| |
Collapse
|
8
|
Lee H, Kang J, Lee SH, Lee D, Chung CH, Lee J. Neuroprotective role of Hippo signaling by microtubule stability control in Caenorhabditis elegans. eLife 2025; 13:RP102001. [PMID: 40178516 PMCID: PMC11968107 DOI: 10.7554/elife.102001] [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] [Indexed: 04/05/2025] Open
Abstract
The evolutionarily conserved Hippo (Hpo) pathway has been shown to impact early development and tumorigenesis by governing cell proliferation and apoptosis. However, its post-developmental roles are relatively unexplored. Here, we demonstrate its roles in post-mitotic cells by showing that defective Hpo signaling accelerates age-associated structural and functional decline of neurons in Caenorhabditis elegans. Loss of wts-1/LATS, the core kinase of the Hpo pathway, resulted in premature deformation of touch neurons and impaired touch responses in a yap-1/YAP-dependent manner, the downstream transcriptional co-activator of LATS. Decreased movement as well as microtubule destabilization by treatment with colchicine or disruption of microtubule-stabilizing genes alleviated the neuronal deformation of wts-1 mutants. Colchicine exerted neuroprotective effects even during normal aging. In addition, the deficiency of a microtubule-severing enzyme spas-1 also led to precocious structural deformation. These results consistently suggest that hyper-stabilized microtubules in both wts-1-deficient neurons and normally aged neurons are detrimental to the maintenance of neuronal structural integrity. In summary, Hpo pathway governs the structural and functional maintenance of differentiated neurons by modulating microtubule stability, raising the possibility that the microtubule stability of fully developed neurons could be a promising target to delay neuronal aging. Our study provides potential therapeutic approaches to combat age- or disease-related neurodegeneration.
Collapse
Affiliation(s)
- Hanee Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| | - Junsu Kang
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| | - Sang-Hee Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| | - Dowoon Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| | - Christine H Chung
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| | - Junho Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
9
|
Elkotamy MS, Abdelrahman MA, Giovannuzzi S, Alkabbani MA, Nocentini A, Supuran CT, Eldehna WM, Abdel-Aziz HA, Abou-Seri SM. Development of pyrazolo[1,5-a]pyrimidine-grafted coumarins as selective carbonic anhydrase inhibitors and tubulin polymerization inhibitors with potent anticancer activity. Int J Biol Macromol 2025; 303:140462. [PMID: 39884639 DOI: 10.1016/j.ijbiomac.2025.140462] [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: 11/19/2024] [Revised: 12/30/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
This study presents the design, synthesis, and evaluation of a novel series of coumarin-based compounds (9a-t) as potential anticancer agents. The compounds were strategically designed to inhibit cancer-related carbonic anhydrase (CA) isoforms IX and XII and tubulin polymerization. Two approaches were employed for CA inhibition: utilizing the coumarin motif to occlude the CA active site entrance and incorporating zinc-binding groups (sulfonamide, carboxylic acid, and thiol) to interact with the catalytic zinc ion. The target compounds were also designed to inhibit tubulin polymerization by combining the privileged coumarin and pyrazolo[1,5-a]pyrimidine scaffolds. Biological evaluation of the target compounds (9a-t) revealed that sulfonamide-containing derivatives 9h and 9r exhibited potent inhibitory activity in the low nanomolar range against CA IX (Ki = 23 and 14 nM, respectively) and CA XII (Ki = 6 and 17 nM, respectively). In NCI-60 human tumor cell line screening, compounds 9k, 9m, and 9q demonstrated broad-spectrum anti-proliferative activity in the five-dose assay with MG-MID values of 7.31 μM, 10.68 μM, and 5.92 μM, respectively. Compound 9m showed significant tubulin polymerization inhibition with an IC50 = 5.28 μM, surpassing the efficacy of colchicine. Cell cycle analysis in MDA-MB-231 breast cancer cells revealed G2/M phase arrest for 9m, which induced significant apoptosis and modulated apoptotic markers. Molecular docking studies provided insights into the binding modes of the compounds with CA IX, CA XII, and tubulin. ADMET and toxicity predictions were performed to assess the drug-like properties of the compounds. These findings pave the way for further optimization of the coumarin scaffold to develop dual inhibitors of carbonic anhydrase IX/XII and tubulin polymerization.
Collapse
Affiliation(s)
- Mahmoud S Elkotamy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo 11829, Egypt
| | - Mohamed A Abdelrahman
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo 11829, Egypt; Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
| | - Simone Giovannuzzi
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Firenze, Italy
| | - Mahmoud Abdelrahman Alkabbani
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo 11829, Egypt
| | - Alessio Nocentini
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Firenze, Italy
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Firenze, Italy
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt.
| | - Hatem A Abdel-Aziz
- Department of Applied Organic Chemistry, National Research Center, Dokki, Cairo 12622, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia Street, Alexandria 21648, Egypt.
| | - Sahar M Abou-Seri
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt.
| |
Collapse
|
10
|
Williams KJ. Inflammation in atherosclerosis: a Big Idea that has underperformed so far. Curr Opin Lipidol 2025; 36:78-87. [PMID: 39846349 PMCID: PMC11888836 DOI: 10.1097/mol.0000000000000973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
PURPOSE OF REVIEW For many years, inflammation has been a major concept in basic research on atherosclerosis and in the development of potential diagnostic tools and treatments. The purpose of this review is to assess the performance of this concept with an emphasis on recent clinical trials. In addition, contemporary literature may help identify new therapeutic targets, particularly in the context of the treatment of early, rather than end-stage, arterial disease. RECENT FINDINGS Newly reported clinical trials cast doubt on the efficacy of colchicine, the sole anti-inflammatory agent currently approved for use in patients with atherosclerotic cardiovascular disease (ASCVD). New analyses also challenge the hypothesis that residual ASCVD event risk after optimal management of lipids, blood pressure, and smoking arises primarily from residual inflammatory risk. Current clinical practice to initiate interventions so late in the course of atherosclerotic arterial disease may be a better explanation. Lipid-lowering therapy in early atherosclerosis, possibly combined with novel add-on agents to specifically accelerate resolution of maladaptive inflammation, may be more fruitful than the conventional approach of testing immunosuppressive strategies in end-stage arterial disease. Also discussed is the ongoing revolution in noninvasive technologies to image the arterial wall. These technologies are changing screening, diagnosis, and treatment of atherosclerosis, including early and possibly reversable disease. SUMMARY The burden of proof that the Big Idea of inflammation in atherosclerosis has clinical value remains the responsibility of its advocates. This responsibility requires convincing trial data but still seems largely unmet. Unfortunately, the focus on inflammation as the source of residual ASCVD event risk has distracted us from the need to screen and treat earlier.
Collapse
Affiliation(s)
- Kevin Jon Williams
- Department of Cardiovascular Sciences and Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Lal S, Snape TJ. Tubulin targeting agents and their implications in non-cancer disease management. Drug Discov Today 2025; 30:104338. [PMID: 40118444 DOI: 10.1016/j.drudis.2025.104338] [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: 07/26/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Microtubules act as molecular 'tracks' for the intracellular transport of accessory proteins, enabling them to assemble into various larger structures, such as spindle fibres formed during the cell cycle. Microtubules provide an organisational framework for the healthy functioning of various cellular processes that work through the process of dynamic instability, driven by the hydrolysis of GTP. In this role, tubulin proteins undergo various modifications, and in doing so modulate various healthy or pathogenic physiological processes within cells. In this review, we provide a detailed update of small molecule chemical agents that interact with tubulin, along with their implications, specifically in non-cancer disease management.
Collapse
Affiliation(s)
- Samridhi Lal
- Amity Institute of Pharmacy, Amity University, Gurugram 122413 Haryana, India.
| | - Timothy J Snape
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| |
Collapse
|
12
|
Liang RS, Su JQ, Wu XQ, Wang Q, Cai YM, Su HY, Tang JX, Yao CW. Identification of therapeutic targets for chronic kidney disease through Mendelian randomization analysis of druggable genes. Sci Rep 2025; 15:10779. [PMID: 40155631 PMCID: PMC11953390 DOI: 10.1038/s41598-025-94761-0] [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: 09/14/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Chronic Kidney Disease (CKD) is a multifaceted and gradually advancing condition characterized by a complex pathogenesis. The current therapeutic options for CKD remain limited in efficacy. Consequently, the identification and exploration of novel drug targets for CKD are of paramount importance. We identified cis-expression quantitative trait loci (cis-eQTLs) with potential as drug targets from the eQTLGen Consortium database to serve as the exposure. For the outcome, we utilized a genome-wide association study (GWAS) of chronic kidney disease (CKD) from the FinnGen database, which comprised a case group of 11,265 individuals and a control group of 436,208 individuals. MR analysis was employed to investigate druggable genes closely associated with CKD. Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted to elucidate the functional roles of these significant genes. Finally, a colocalization analysis was conducted to determine the likelihood that a cis-eQTL for a druggable gene and CKD share a causal variant. The expression of 12 genes was found to be significantly associated with CKD risk, with a false discovery rate (FDR) of less than 0.05. GO and KEGG enrichment analyses indicated that these genes are primarily involved in the regulation of MAP kinase activity, regulation of protein serine/threonine kinase activity, Gap junction, Platelet activation and Oxytocin signaling pathway. The colocalization analysis results suggested that CKD and the TUBB gene may share a causal variant, with a posterior probability (PP.H4) exceeding 80% (TUBB: 97.27%). Compelling statistical evidence indicates that TUBB represents the most promising pharmacological target for the treatment of CKD. This study not only identifies potential therapeutic targets but also offers valuable insights for future drug development in the context of CKD.
Collapse
Affiliation(s)
- Run-Sen Liang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jin-Qi Su
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiang-Qi Wu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qi Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yong-Mei Cai
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Hong-Yong Su
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Ji-Xin Tang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Cui-Wei Yao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
13
|
Enneiymy M, El Aissouq A. Carvacrol-Derived 1,2,3-Triazole Hybrids: Synthesis, Computational Insights, and Targeted Inhibition of EGFR, BRAF V600E, and Tubulin Enzymes. J Fluoresc 2025:10.1007/s10895-025-04232-y. [PMID: 40100316 DOI: 10.1007/s10895-025-04232-y] [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: 01/11/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
This study explores the design and synthesis of innovative triazole-carvacrol hybrid molecules via copper-catalyzed 1,3-dipolar cycloaddition reactions. Leveraging advanced computational drug design tools, the ADMET (absorption, distribution, metabolism, excretion, and toxicity) profiles of these compounds will be meticulously evaluated. Furthermore, molecular docking simulations will unravel the binding interactions and mechanisms with critical cancer therapy targets, including EGFR (PDB ID: 3POZ), BRAF V600E (PDB ID: 1UWJ), and Tubulin (PDB ID: 1SA0). By integrating cutting-edge synthesis and computational techniques, this work aims to uncover potent candidates with significant therapeutic potential in cancer treatment.
Collapse
Affiliation(s)
- Mohamed Enneiymy
- Laboratory of Organic and Physical Chemistry, Applied Bioorganic Chemistry Team, Faculty of Sciences, Ibnou Zohr University, Agadir, Morocco.
| | - Abdellah El Aissouq
- Laboratory of Processes, Materials, and Environment (LPME), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Fez, Morocco.
| |
Collapse
|
14
|
Li Y, Zhang C, Tang D, Wang T, Yan W, Yang L, Bai P, Tang M, Pei H, Chen L, Chen Q, Yang J. Identification of a ligand-binding site on tubulin mediating the tubulin-RB3 interaction. Proc Natl Acad Sci U S A 2025; 122:e2424098122. [PMID: 40067895 PMCID: PMC11929442 DOI: 10.1073/pnas.2424098122] [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: 11/19/2024] [Accepted: 01/29/2025] [Indexed: 03/17/2025] Open
Abstract
For decades, microtubules-composed of αβ-tubulin dimers-have been primary targets for cancer chemotherapy. While eight binding sites on the tubulin dimer have been structurally characterized, this study reveals a ninth. We found that the tubulin inhibitor Tumabulin-1 (TM1, a BML284 derivative) binds simultaneously to the well-known colchicine site and a previously unknown site, designated as Tumabulin site. This site resides at the interface of α1-tubulin, β1-tubulin, and RB3 within the tubulin-RB3-tubulintyrosine ligase complex. Remarkably, two TM1 molecules bind cooperatively to this relatively large pocket, interacting with all three proteins. Crucially, this binding is dependent on RB3; it is absent when RB3 is missing or the key residue H71 is mutated (H71Q). We further designed and synthesized Tumabulin-2 (TM2) that selectively binds the Tumabulin site, excluding binding the colchicine site. TM2 acts as a molecular glue, strengthening the interaction between RB3 and the tubulin dimer and consequently enhancing RB3's tubulin-depolymerizing activity. In conclusion, our findings confirm the existence of a ninth tubulin-binding site and offer a promising foundation for developing Tubulin-RB3 molecular glues as a next generation of anticancer therapeutics.
Collapse
Affiliation(s)
- Yong Li
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan610041, China
| | - Chufeng Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Dongmei Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Tao Wang
- Natural and Biomimetic Medicine Research Center, Tissue-Orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Wei Yan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Linyu Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Peng Bai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Heying Pei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Lijuan Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Qiang Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Jianhong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| |
Collapse
|
15
|
Rubicondo M, Ciardelli G, Mattu C, Tuszynski JA. Recent advancements in colchicine derivatives: Exploring synthesis, activities, and nanoformulations for enhanced therapeutic efficacy. Drug Discov Today 2025; 30:104312. [PMID: 39947582 DOI: 10.1016/j.drudis.2025.104312] [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: 11/07/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
The multifaceted anti-cancer properties of colchicine make it a promising candidate for tumor treatment. However, its application has been limited by poor solubility, low bioavailability, and systemic toxicity. Considerable efforts have been directed toward the development of colchicine derivatives and nanoformulations to overcome these challenges. In this review, we provide a comprehensive overview of recent advances in colchicine derivatives and nanoformulations for cancer treatment. Synthesis methods and in vitro antiproliferative assays for the reviewed derivatives and formulations are explored. Challenges, such as drug resistance and formulation optimization, are also addressed, along with future perspectives for leveraging the full potential of colchicine derivatives and their nanoformulations as innovative anti-cancer strategies, toward successful clinical applications.
Collapse
Affiliation(s)
- Marialucia Rubicondo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Polito BIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Polito BIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Polito BIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Jack A Tuszynski
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada.
| |
Collapse
|
16
|
Jin L, Qin Y, Zhao Y, Zhou X, Zeng Y. Endothelial cytoskeleton in mechanotransduction and vascular diseases. J Biomech 2025; 182:112579. [PMID: 39938443 DOI: 10.1016/j.jbiomech.2025.112579] [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: 11/27/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The cytoskeleton is an important structural component that regulates various aspects of cell morphology, movement, and intracellular signaling. It plays a pivotal role in the cellular response to biomechanical stimuli, particularly in endothelial cells, which are critical for vascular homeostasis and the pathogenesis of cardiovascular diseases. Mechanical forces, such as shear and tension, activate intracellular signaling cascades that regulate transcription, translation, and cellular behaviors. Despite extensive research into cytoskeletal functions, the precise mechanisms by which the cytoskeleton transduces mechanical signals remain incompletely understood. This review focuses on the role of cytoskeletal components in membrane, cytoplasm, and nucleus in mechanotransduction, with an emphasis on their structure, mechanical and biological behaviors, dynamic interactions, and response to mechanical forces. The collaboration between membrane cytoskeleton, cytoplasmic cytoskeleton, and nucleoskeleton is indispensable for endothelial cells to respond to mechanical stimuli. Understanding their mechanoresponsive mechanisms is essential for advancing therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Linlu Jin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yixue Qin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yunran Zhao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Xintong Zhou
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China.
| |
Collapse
|
17
|
Al-Hawamdeh MI, Othman F, Taha S, Adawı T, Aburjaı T. Insights into familial Mediterranean fever: Chronic disease correlations with arthralgia and current health status of patients with familial Mediterranean fever in Jordan. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2025; 6:21-28. [PMID: 40191470 PMCID: PMC11966200 DOI: 10.1515/rir-2025-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/16/2024] [Indexed: 04/09/2025]
Abstract
Background and Objectives Familial Mediterranean fever (FMF) stands as a significant challenge within Jordan's clinical practice, despite its low prevalence of 0.04% within the Jordanian population. This study aims to investigate the current status of the health status of FMF patients in Jordan while exploring any present associations between chronic diseases and the severity of their symptoms. Methods This is a cross-sectional descriptive survey-based study conducted during the period between 1st of March till the last of May 2023 in Jordan. The survey was distributed randomly to a group of FMF patients, Sample size was based on FMF prevalence in Jordan (0.04%); study sample (N = 67) included FMF patients in Jordan from different age groups. All results were performed through proper statistical analysis. Results The study includes 67 FMF patients, predominantly Jordanian and aged 18-31, revealed that 58.2% only were diagnosed through blood genetic testing. Marriages among first-degree relatives showed a 60% probability of FMF transmission compared to 10% in non-related parents (P = 0.001), leading 82.1% of participants to call for pre-marital testing. Acute symptoms included abdominal pain, fever, arthralgia, and myalgia, with hypertension being the most frequent comorbidity (14.9%) and significantly associated with myalgia and arthralgia (P < 0.05). Colchicine was the primary treatment for 89.6% of patients, with high adherence rates (90.3%). Conclusion Among chronic comorbidities, hypertension was associated in increasing the severity of the myalgia during attacks. The issue of misdiagnosis remains a major challenge in Jordanian clinical practice. Our findings assert the importance of future incorporation of FMF premarital testing.
Collapse
Affiliation(s)
- Mai I. Al-Hawamdeh
- Department of pharmacy, College of pharmacy, Amman Arab University, Amman, Jordan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University Of Jordan, Amman, Jordan
| | - Farah Othman
- Department of pharmacy, College of pharmacy, Amman Arab University, Amman, Jordan
| | - Safaa’ Taha
- Department of pharmacy, College of pharmacy, Amman Arab University, Amman, Jordan
| | - Tityana Adawı
- Department of pharmacy, College of pharmacy, Amman Arab University, Amman, Jordan
| | - Talal Aburjaı
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University Of Jordan, Amman, Jordan
| |
Collapse
|
18
|
Deng X, Bradshaw G, Kalocsay M, Mitchison T. Tubulin Regulates the Stability and Localization of STMN2 by Binding Preferentially to Its Soluble Form. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640326. [PMID: 40060442 PMCID: PMC11888388 DOI: 10.1101/2025.02.27.640326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Loss of the tubulin-binding protein STMN2 is implicated in amyotrophic lateral sclerosis (ALS) but how it protects neurons is not known. STMN2 is known to turn over rapidly and accumulate at axotomy sites. We confirmed fast turnover of STMN2 in U2OS cells and iPSC-derived neurons and showed that degradation occurs mainly by the ubiquitin-proteasome system. The membrane targeting N-terminal domain of STMN2 promoted fast turnover, whereas its tubulin binding stathmin-like domain (SLD) promoted stabilization. Proximity labeling and imaging showed that STMN2 localizes to trans-Golgi network membranes and that tubulin binding reduces this localization. Pull-down assays showed that tubulin prefers to bind to soluble over membrane-bound STMN2. Our data suggest that STMN2 interconverts between a soluble form that is rapidly degraded unless bound to tubulin and a membrane-bound form that does not bind tubulin. We propose that STMN2 is sequestered and stabilized by tubulin binding, while its neuroprotective function depends on an unknown molecular activity of its membrane-bound form.
Collapse
Affiliation(s)
- Xiang Deng
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Gary Bradshaw
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Marian Kalocsay
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Timothy Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
19
|
Tran C, Hamze A. Recent Advancements in the Development of HDAC/Tubulin Dual-Targeting Inhibitors. Pharmaceuticals (Basel) 2025; 18:341. [PMID: 40143119 PMCID: PMC11945613 DOI: 10.3390/ph18030341] [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: 01/14/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Histone deacetylases (HDACs) have become one of the main targets in cancer therapy due to their involvement in various biological processes, including gene regulation, cell proliferation, and differentiation. Microtubules, as key elements of the cell cytoskeleton, also represent important therapeutic targets in anticancer drugs research. These proteins are involved in diverse cellular functions, especially mitosis, cell signaling, and intracellular trafficking. With the emergence of multi-target therapy during the last decades, the combination of HDAC and tubulin inhibitors has been envisioned as a practical approach for optimizing the therapeutic efficacy of antitumor molecules. HDAC/tubulin dual-targeting inhibitors offer the advantages of the synergistic action of both compounds, along with a significant decrease in their respective toxicities and drug resistance. This review will detail the major recent advancements in the development of HDAC/tubulin dual inhibitors over the last decade and their impact on anticancer drugs discovery.
Collapse
Affiliation(s)
- Christine Tran
- BioCIS, CNRS (Centre National de Recherche Scientifique), Université Paris-Saclay, 91400 Orsay, France
| | - Abdallah Hamze
- BioCIS, CNRS (Centre National de Recherche Scientifique), Université Paris-Saclay, 91400 Orsay, France
| |
Collapse
|
20
|
Vairin R, Tamminga C, Shi Z, Borchardt C, Jambulapati J, Bai R, Wanniarachchi H, Bueno L, Hamel E, Mason RP, Trawick ML, Pinney KG. Design, Synthesis and Biological Evaluation of 2-Phenyl Indole Analogues of OXi8006 as Colchicine Site Inhibitors of Tubulin Polymerization and Vascular Disrupting Agents. Bioorg Med Chem 2025; 118:117981. [PMID: 39667146 PMCID: PMC11834100 DOI: 10.1016/j.bmc.2024.117981] [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: 09/20/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 12/14/2024]
Abstract
Inhibitors of tubulin polymerization represent a promising therapeutic approach for the treatment of solid tumors. Molecules that bind to the colchicine site are of interest as they can function with a dual mechanism of action as both potent antiproliferative agents and tumor-selective vascular disrupting agents (VDAs). One such example is a 2-aryl-3-aroyl-indole molecule (OXi8006) from our laboratory that demonstrates potent inhibition of tubulin polymerization and strong antiproliferative activity (cytotoxicity) against a variety of human cancer cell lines. A water-soluble prodrug OXi8007, synthesized from OXi8006, demonstrates in vivo disruption of tumor-associated microvessels in several tumor types (mouse models). The molecular framework of OXi8006 inspired a series of fourteen new 2-aryl-3-aroyl-indole analogues that incorporated various functional group modifications on both the indole core and the aroyl ring. Electron withdrawing and donating groups at the mono-substituted 3' position and the di-substituted 3',5' positions were all accommodated while maintaining inhibition of tubulin polymerization (IC50 < 5 μM), with several analogues demonstrating activity comparable to OXi8006 and the benchmark natural product combretastatin A-4 (CA4). Preliminary structure-activity relationship (SAR) studies were further enhanced by molecular docking to predict possible colchicine site interactions. Two analogues (KGP366 and KGP369) previously synthesized in our laboratory were re-synthesized using a somewhat modified route to increase synthetic efficiency and were subsequently converted to their corresponding water-soluble phosphate prodrug salts to evaluate their efficacy as VDAs. Administration of the prodrug salt (KGP415) of KGP369 caused significant reduction in bioluminescence signal from an orthotopic kidney tumor (RENCA-luc) in BALB/c mice, indicative of VDA activity. Collectively, these new functionalized indole-based analogues have extended SAR knowledge related to the colchicine binding site, and the most biologically active analogues hold promise for continued development as pre-clinical candidates for cancer therapy.
Collapse
Affiliation(s)
- Rebecca Vairin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Caleb Tamminga
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Zhe Shi
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Christian Borchardt
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Jayaram Jambulapati
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Hashini Wanniarachchi
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - Lorena Bueno
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Ralph P Mason
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States.
| |
Collapse
|
21
|
Strus P, Sadowski K, Ploch W, Jazdzewska A, Oknianska P, Raniszewska O, Mlynarczuk-Bialy I. The Effects of Podophyllotoxin Derivatives on Noncancerous Diseases: A Systematic Review. Int J Mol Sci 2025; 26:958. [PMID: 39940726 PMCID: PMC11816842 DOI: 10.3390/ijms26030958] [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: 12/26/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Podophyllotoxin (PPT) is commonly used for genital warts due to its antimitotic properties and relatively good accessibility since it can be extracted from plants in low-economy countries. However, due to relatively high toxicity, it cannot be used in a systematic way (intravenously). Thus, there is a need to find or create an equally effective derivative of PPT that will be less toxic. Natural PPT is a suitable and promising scaffold for the synthesis of its derivatives. Many of them have been studied in clinical and preclinical models. In this systematic review, we comprehensively assess the medical applications of PPT derivatives, focusing on their advantages and limitations in non-cancerous diseases. Most of the existing research focuses on their applications in cancerous diseases, leaving non-cancerous uses underexplored. To do that, we systematically reviewed the literature using PubMed, Embase, and Cochrane databases from January 2013 to January 2025. In total, 5333 unique references were identified in the initial search, of which 44 were included in the quantitative synthesis. The assessment of the quality of eligible studies was undertaken using the PRISMA criteria. The risk of bias was assessed using a predefined checklist based on PRISMA guidelines. Each study was independently reviewed by two researchers to evaluate bias in study design, reporting, and outcomes. Our analysis highlights the broad therapeutic potential of PPT derivatives, particularly in antiviral applications, including HPV, Dengue, and SARS-CoV-2 infections. Apart from their well-known anti-genital warts activity, these compounds exhibit significant anti-inflammatory, antimitotic, analgesic, and radioprotective properties. For instance, derivatives such as cyclolignan SAU-22.107 show promise in antiviral therapies, while compounds like G-003M demonstrate radioprotective effects by mitigating radiation-induced damage. To build on this, our review highlights that PPT derivatives, apart from anti-genital warts potential, exhibit four key properties-anti-inflammatory, antimitotic, analgesic, and radioprotective-making them promising candidates not only for treating viral infections such as HPV, Dengue, and SARS-CoV-2 but also for expanding their therapeutic potential beyond cancerous diseases. In conclusion, while PPT derivatives hold great potential across various medical domains, their applications in non-cancerous diseases remain limited by the scarcity of dedicated research. Continued exploration of these compounds is essential to unlock their full therapeutic value.
Collapse
Affiliation(s)
- Piotr Strus
- Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.); (W.P.)
| | - Karol Sadowski
- Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.); (W.P.)
| | - Weronika Ploch
- Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.); (W.P.)
| | - Adrianna Jazdzewska
- Student Scientific Circle of Rare Diseases at Department of Pediatrics, Hematology and Oncology, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Paulina Oknianska
- Student Scientific Circle of Oncology and Radiotherapy at Department of Oncology and Radiotherapy, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Oliwia Raniszewska
- Student Scientific Circle of Child and Adolescent Psychiatry, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Izabela Mlynarczuk-Bialy
- Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.); (W.P.)
| |
Collapse
|
22
|
Ana G, Malebari AM, Noorani S, Fayne D, O’Boyle NM, Zisterer DM, Pimentel EF, Endringer DC, Meegan MJ. ( E)-1-(3-(3-Hydroxy-4-Methoxyphenyl)-1-(3,4,5-Trimethoxyphenyl)allyl)-1 H-1,2,4-Triazole and Related Compounds: Their Synthesis and Biological Evaluation as Novel Antimitotic Agents Targeting Breast Cancer. Pharmaceuticals (Basel) 2025; 18:118. [PMID: 39861179 PMCID: PMC11769294 DOI: 10.3390/ph18010118] [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: 11/24/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
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 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.
Collapse
Affiliation(s)
- Gloria Ana
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Azizah M. Malebari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sara Noorani
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Darren Fayne
- Molecular Design Group, School of Chemical Sciences, Dublin City University, Glasnevin, D09 V209 Dublin, Ireland
- DCU Life Sciences Institute, Dublin City University, Glasnevin, D09 V209 Dublin, Ireland
| | - Niamh M. O’Boyle
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Daniela M. Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, D02 R590 Dublin, Ireland
| | - Elisangela Flavia Pimentel
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista, Vila Velha CEP 29102-920, Brazil
| | - Denise Coutinho Endringer
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista, Vila Velha CEP 29102-920, Brazil
| | - Mary J. Meegan
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| |
Collapse
|
23
|
Adekunle YA, Samuel BB, Ezeude CM, Nahar L, Fatokun AA, Sarker SD. Isolation, cytotoxicity evaluation, and molecular docking of 3,4,3'-tri- O-methylflavellagic acid from Anogeissus leiocarpus (DC.) Guill. & Perr. root. Nat Prod Res 2025:1-8. [PMID: 39798146 DOI: 10.1080/14786419.2025.2451218] [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: 05/24/2024] [Revised: 12/11/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Cancer kills about 10 million people every year. Medicinal plants remain a major source in the global search for anticancer drugs. In this study, 3,4,3'-tri-O-methylflavellagic acid (MFA) was isolated from the methanol root extract of Anogeissus leiocarpus. The structure was determined by 1D- and 2D-NMR data. The cytotoxic effects of MFA were evaluated against human breast (MCF-7), colorectal (Caco-2), and cervical (HeLa) cancer cell lines using the 3-[4,5-dimethylthiazole-2-yl] 3,5-diphenyltetrazolium bromide assay. A multi-protein target screening via molecular docking was conducted against ten cancer-related proteins, and ADMET properties were evaluated. MFA exhibited the most potent activity against Caco-2 (IC50: 46.75 ± 13.00 µM). Molecular docking analysis showed that MFA had a strong binding affinity for the colchicine-binding site of αβ-tubulin and polo-like kinase-1 (binding energies: -8.5 and -8.4 kcal/mol, respectively). MFA also satisfied the Lipinski's Rule of Five. MFA could, therefore, potentially serve as a scaffold for developing new anticancer molecules.
Collapse
Affiliation(s)
- Yemi A Adekunle
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
- Department of Pharmaceutical Chemistry, Dora Akunyili College of Pharmacy, Igbinedion University, Benin City, Edo State, Nigeria
| | - Babatunde B Samuel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Chinemenma M Ezeude
- Department of Pharmaceutical Chemistry, Dora Akunyili College of Pharmacy, Igbinedion University, Benin City, Edo State, Nigeria
| | - Lutfun Nahar
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czech Republic
| | - Amos A Fatokun
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Satyajit D Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
24
|
Yurtsever A, Asakawa H, Katagiri Y, Takao K, Ikegami K, Tsukada M, Setou M, Fukuma T. Visualizing the Submolecular Organization of αβ-Tubulin Subunits on the Microtubule Inner Surface Using Atomic Force Microscopy. NANO LETTERS 2025; 25:98-105. [PMID: 39569635 DOI: 10.1021/acs.nanolett.4c04294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Microtubules (MTs) are dynamic cytoskeletal polymers essential for mediating fundamental cellular processes, including cell division, intracellular transport, and cell shape maintenance. Understanding the arrangement of tubulin heterodimers within MTs is key to their function. Using frequency modulation atomic force microscopy (FM-AFM) and simulations, we revealed the submolecular arrangement of α- and β-tubulin subunits on the inner MT surface. We observed an undulating molecular arrangement of protofilaments (PFs) with alternating height variations, attributed to different structural orientations and the confirmation of αβ-tubulin heterodimers in adjacent PFs, forming bimodal lateral contacts, as confirmed by AFM simulations. Structural defects resulting from missing tubulin units were directly identified. This detailed structural information provides critical insight into the MT functional properties. Our findings highlight the potential of FM-AFM in liquid as a powerful tool for elucidating the complex interactions among MTs, MT-associated proteins, and other molecules, which are essential for understanding MT dynamics in the cellular context.
Collapse
Affiliation(s)
- Ayhan Yurtsever
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Hitoshi Asakawa
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
- Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa 920-1192, Japan
- Nanomaterials Research Institute (NanoMaRi), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yukitoshi Katagiri
- Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa 920-1192, Japan
| | - Kazufumi Takao
- Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa 920-1192, Japan
| | - Koji Ikegami
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Masaru Tsukada
- WPI Advanced Institute for Materials Research, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai 980-8577, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- International Mass Imaging and Spatial Omics Center, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Takeshi Fukuma
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
- Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa 920-1192, Japan
| |
Collapse
|
25
|
Hernández Á, Rosales‐Fernández C, Miranda‐Vera C, Veselinova A, Jambrina PG, García‐García P, García PA, Díez D, Castro MÁ, Fuentes M. Insights into podophyllotoxin lactone features: New cyclolignans as potential dual tubulin-topoisomerase II inhibitors. Arch Pharm (Weinheim) 2025; 358:e2400600. [PMID: 39533473 PMCID: PMC11726159 DOI: 10.1002/ardp.202400600] [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: 07/23/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Chemomodulation of natural cyclolignans as podophyllotoxin has been a successful approach to obtain semisynthetic bioactive derivates. One example of this approach is the FDA-approved drug etoposide for solid and hematological tumors. It differs from the antimitotic activity of the natural product in its mechanism of action, this drug being a topoisomerase II inhibitor instead of a tubulin antimitotic. Within the molecular requirements for the activity of these compounds, the trans-γ-lactone moiety presented in the parent compound has always been a feature to be explored to chemomodulate its bioactivity. In this study, we have obtained different compounds that comply with the molecular characteristics for antitubulin and antitopoisomerase II activity combined in a single molecule. Furthermore, we explored the influence of the trans-lactone moiety on the final activity, finding that the cis-lactone was also interesting in terms of bioactivity. The best values of cytotoxicity and cell cycle inhibition were obtained for a compound lacking the lactone ring, thus mimicking the podophyllic aldehyde functionalization, a selective antimitotic podophyllotoxin derivate. The analogs with cis-lactone also presented interesting cytotoxic activity. The present study illustrates the potential of the chemomodulation of natural products such as natural cyclolignan podophyllotoxin derivates for the discovery of new antitumor agents.
Collapse
Affiliation(s)
- Ángela‐Patricia Hernández
- Laboratorio de Química Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Campus Miguel de UnamunoUniversidad de SalamancaSalamancaSpain
- Department of Medicine and General Cytometry Service‐Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), IBSAL, Campus Miguel de UnamunoUniversidad de Salamanca‐CSICSalamancaSpain
| | - Celia Rosales‐Fernández
- Laboratorio de Química Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Campus Miguel de UnamunoUniversidad de SalamancaSalamancaSpain
| | - Carolina Miranda‐Vera
- Laboratorio de Química Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Campus Miguel de UnamunoUniversidad de SalamancaSalamancaSpain
| | - Anzhela Veselinova
- Departamento de Química Física, Facultad de Ciencias QuímicasUniversidad de SalamancaSalamancaSpain
| | - Pablo G. Jambrina
- Departamento de Química Física, Facultad de Ciencias QuímicasUniversidad de SalamancaSalamancaSpain
| | - Pilar García‐García
- Laboratorio de Química Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Campus Miguel de UnamunoUniversidad de SalamancaSalamancaSpain
| | - Pablo A. García
- Laboratorio de Química Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Campus Miguel de UnamunoUniversidad de SalamancaSalamancaSpain
| | - David Díez
- Departamento de Química Orgánica, Facultad de Ciencias QuímicasUniversidad de SalamancaSalamancaSpain
| | - María Ángeles Castro
- Laboratorio de Química Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Campus Miguel de UnamunoUniversidad de SalamancaSalamancaSpain
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service‐Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), IBSAL, Campus Miguel de UnamunoUniversidad de Salamanca‐CSICSalamancaSpain
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| |
Collapse
|
26
|
Zhu L, Zhang M, Leng J, Zhao B, Ning M, Zhang C, Kong L, Yin Y. Discovery of novel quinazoline derivatives as tubulin polymerization inhibitors targeting the colchicine binding site with potential anti-colon cancer effects. Eur J Med Chem 2024; 280:117000. [PMID: 39489984 DOI: 10.1016/j.ejmech.2024.117000] [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: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Tubulin is a critical target for cancer therapy, with colchicine binding site inhibitors (CBSIs) being the most extensively researched. A series of quinazoline derivatives designed to target the colchicine binding site of tubulin were synthesized and evaluated for their biological activities. The antiproliferative effects of these compounds were tested against six human cancer cell lines, and compound Q19 demonstrated potent antiproliferative activity against the HT-29 cell line, with an IC50 value of 51 nM. Additionally, further investigation revealed that Q19 effectively inhibited microtubule polymerization by binding to the colchicine binding site on tubulin. Furthermore, compound Q19 arrested the HT-29 cell cycle at the G2/M phase, induced apoptosis in these cells, and disrupted angiogenesis. Finally, compound Q19 exhibited potent inhibitory effects on tumor growth in HT-29 xenografted mice while demonstrating minimal toxic side effects and acceptable pharmacokinetic properties. These findings suggested that Q19 hold promise as a potential candidate for colon cancer therapy targeting tubulin.
Collapse
Affiliation(s)
- Liqiao Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Mengyu Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Jiafu Leng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Bo Zhao
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Mengdan Ning
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Chao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| | - Yong Yin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
27
|
Tenorio JCB, Heikal MF, Kafle A, Saichua P, Suttiprapa S. Benzimidazole resistance-associated mutations improve the in silico dimerization of hookworm tubulin: An additional resistance mechanism. Vet World 2024; 17:2736-2746. [PMID: 39897360 PMCID: PMC11784061 DOI: 10.14202/vetworld.2024.2736-2746] [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: 08/16/2024] [Accepted: 11/07/2024] [Indexed: 02/04/2025] Open
Abstract
Background and Aim Mutations in the β-tubulin genes of helminths confer benzimidazole (BZ) resistance by reducing the drug's binding efficiency to the expressed protein. However, the effects of these resistance-associated mutations on tubulin dimer formation in soil-transmitted helminths remain unknown. Therefore, this study aimed to investigate the impact of these mutations on the in silico dimerization of hookworm α- and β-tubulins using open-source bioinformatics tools. Materials and Methods Using AlphaFold 3, the α- and β-tubulin amino acid sequences of Ancylostoma ceylanicum were used to predict the structural fold of the hookworm tubulin heterodimer. The modeled complexes were subjected to several protein structure quality assurance checks. The binding free energies, overall binding affinity, dissociation constant, and interacting amino acids of the complex were determined. The dimer's structural flexibility and motion were simulated through molecular dynamics. Results BZ resistance-associated amino acid substitutions in the β-tubulin isotype 1 protein of hookworms altered tubulin dimerization. The E198K, E198V, and F200Y mutations conferred the strongest and most stable binding between the α and β subunits, surpassing that of the wild-type. In contrast, complexes with the Q134H and F200L mutations exhibited the opposite effect. Molecular dynamics simulations showed that wild-type and mutant tubulin dimers exhibited similar dynamic behavior, with slight deviations in those carrying the F200L and E198K mutations. Conclusion Resistance-associated mutations in hookworms impair BZ binding to β-tubulin and enhance tubulin dimer interactions, thereby increasing the parasite's ability to withstand treatment. Conversely, other mutations weaken these interactions, potentially compromising hookworm viability. These findings offer novel insights into helminth tubulin dimerization and provide a valuable foundation for developing anthelmintics targeting this crucial biological process.
Collapse
Affiliation(s)
- Jan Clyden B. Tenorio
- Tropical Medicine Graduate Program, Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Veterinary Paraclinical Sciences, College of Veterinary Medicine, University of Southern Mindanao, Kabacan 9407, Cotabato, Philippines
| | - Muhammad Fikri Heikal
- Tropical Medicine Graduate Program, Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Alok Kafle
- Tropical Medicine Graduate Program, Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Prasert Saichua
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- WHO Collaborating Center for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sutas Suttiprapa
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- WHO Collaborating Center for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
28
|
Wang C, Zhang Y, Yang S, Shi L, Rong R, Zhang T, Wu Y, Xing D. Design, synthesis, and bioevaluation of 1 h-pyrrolo[3,2- c]pyridine derivatives as colchicine-binding site inhibitors with potent anticancer activities. J Enzyme Inhib Med Chem 2024; 39:2302320. [PMID: 38221788 PMCID: PMC10791102 DOI: 10.1080/14756366.2024.2302320] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/29/2023] [Indexed: 01/16/2024] Open
Abstract
A new series of 1H-pyrrolo[3,2-c]pyridine derivatives were designed and synthesised as colchicine-binding site inhibitors. Preliminary biological evaluations showed that most of the target compounds displayed moderate to excellent antitumor activities against three cancer cell lines (HeLa, SGC-7901, and MCF-7) in vitro. Among them, 10t exhibited the most potent activities against three cancer cell lines with IC50 values ranging from 0.12 to 0.21 μM. Tubulin polymerisation experiments indicated that 10t potently inhibited tubulin polymerisation at concentrations of 3 μM and 5 μM, and immunostaining assays revealed that 10t remarkably disrupted tubulin microtubule dynamics at a concentration of 0.12 μM. Furthermore, cell cycle studies and cell apoptosis analyses demonstrated that 10t at concentrations of 0.12 μM, 0.24 μM, and 0.36 μM significantly caused G2/M phase cell cycle arrest and apoptosis. The results of molecular modelling studies suggested that 10t interacts with tubulin by forming hydrogen bonds with colchicine sites Thrα179 and Asnβ349. In addition, the prediction of physicochemical properties disclosed that 10t conformed well to the Lipinski's rule of five.
Collapse
Affiliation(s)
- Chao Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Shanbo Yang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Lingyu Shi
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Rong Rong
- Yantai Key laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, China
| | - Tingting Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
29
|
Chowdhary S, Preeti, Shekhar, Gupta N, Kumar R, Kumar V. Advances in chalcone-based anticancer therapy: mechanisms, preclinical advances, and future perspectives. Expert Opin Drug Discov 2024; 19:1417-1437. [PMID: 39621431 DOI: 10.1080/17460441.2024.2436908] [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: 06/17/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024]
Abstract
INTRODUCTION Cancer remains a leading cause of death worldwide with traditional treatments like chemotherapy, and radiotherapy becoming less effective due to multidrug resistance (MDR). This highlights the necessity for novel chemotherapeutics like chalcone-based compounds, which demonstrate broad anti-cancer properties and target multiple pathways. These compounds hold promise for improving cancer treatment outcomes compared to existing therapies. AREAS COVERED This review provides a comprehensive synopsis of the recent literature (2018-2024) for anti-proliferative/anti-cancer activity of chalcones. It includes the identification of potential targets, their mechanisms of action, and possible modes of binding. Additionally, chalcone derivatives in preclinical trials are also discussed. EXPERT OPINION Chalcones mark a significant stride in anticancer therapies due to their multifaceted approach in targeting various cellular pathways. Their ability to simultaneously target multiple pathways enables them to overcome drug resistance as compared to traditional therapies. With well-defined mechanisms of action, these compounds can serve as lead molecules for designing new, more promising treatments. Continued progress in synthesis and structural optimization, along with promising results from preclinical trials, offers hope for the development of more potent molecules, heralding a new era in cancer therapeutics.
Collapse
Affiliation(s)
| | - Preeti
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Shekhar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Nikita Gupta
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Rajesh Kumar
- Department of Physics, Lovely Professional University, Phagwara, India
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
30
|
Curcio A, Rocca R, Chiera F, Gallo Cantafio ME, Valentino I, Ganino L, Murfone P, De Simone A, Di Napoli G, Alcaro S, Amodio N, Artese A. Hit Identification and Functional Validation of Novel Dual Inhibitors of HDAC8 and Tubulin Identified by Combining Docking and Molecular Dynamics Simulations. Antioxidants (Basel) 2024; 13:1427. [PMID: 39594568 PMCID: PMC11591096 DOI: 10.3390/antiox13111427] [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: 10/25/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Chromatin organization, which is under the control of histone deacetylases (HDACs), is frequently deregulated in cancer cells. Amongst HDACs, HDAC8 plays an oncogenic role in different neoplasias by acting on both histone and non-histone substrates. Promising anti-cancer strategies have exploited dual-targeting drugs that inhibit both HDAC8 and tubulin. These drugs have shown the potential to enhance the outcome of anti-cancer treatments by simultaneously targeting multiple pathways critical to disease onset and progression. In this study, a structure-based virtual screening (SBVS) of 96403 natural compounds was performed towards the four Class I HDAC isoforms and tubulin. Using molecular docking and molecular dynamics simulations (MDs), we identified two molecules that could selectively interact with HDAC8 and tubulin. CNP0112925 (arundinin), bearing a polyphenolic structure, was confirmed to inhibit HDAC8 activity and tubulin organization, affecting breast cancer cell viability and triggering mitochondrial superoxide production and apoptosis.
Collapse
Affiliation(s)
- Antonio Curcio
- Department of Health Sciences, University Magna Græcia, 88100 Catanzaro, Italy; (A.C.); (F.C.); (S.A.); (A.A.)
| | - Roberta Rocca
- Department of Health Sciences, University Magna Græcia, 88100 Catanzaro, Italy; (A.C.); (F.C.); (S.A.); (A.A.)
- Net4Science Srl, University Magna Græcia, 88100 Catanzaro, Italy
- Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per L’innovazione Rurale, Località Condoleo di Belcastro, 88100 Catanzaro, Italy
| | - Federica Chiera
- Department of Health Sciences, University Magna Græcia, 88100 Catanzaro, Italy; (A.C.); (F.C.); (S.A.); (A.A.)
| | - Maria Eugenia Gallo Cantafio
- Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy; (M.E.G.C.); (I.V.); (L.G.); (P.M.)
| | - Ilenia Valentino
- Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy; (M.E.G.C.); (I.V.); (L.G.); (P.M.)
| | - Ludovica Ganino
- Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy; (M.E.G.C.); (I.V.); (L.G.); (P.M.)
| | - Pierpaolo Murfone
- Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy; (M.E.G.C.); (I.V.); (L.G.); (P.M.)
| | - Angela De Simone
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10125 Turin, Italy; (A.D.S.); (G.D.N.)
| | - Giulia Di Napoli
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10125 Turin, Italy; (A.D.S.); (G.D.N.)
| | - Stefano Alcaro
- Department of Health Sciences, University Magna Græcia, 88100 Catanzaro, Italy; (A.C.); (F.C.); (S.A.); (A.A.)
- Net4Science Srl, University Magna Græcia, 88100 Catanzaro, Italy
- Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per L’innovazione Rurale, Località Condoleo di Belcastro, 88100 Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy; (M.E.G.C.); (I.V.); (L.G.); (P.M.)
| | - Anna Artese
- Department of Health Sciences, University Magna Græcia, 88100 Catanzaro, Italy; (A.C.); (F.C.); (S.A.); (A.A.)
- Net4Science Srl, University Magna Græcia, 88100 Catanzaro, Italy
| |
Collapse
|
31
|
Herman J, Vanstreels E, Bardiot D, Prota AE, Gaillard N, Gao LJ, Vercruysse T, Persoons L, Daems T, Waer M, Herdewijn P, Louat T, Steinmetz MO, De Jonghe S, Sprangers B, Daelemans D. 3-nitropyridine analogues as novel microtubule-targeting agents. PLoS One 2024; 19:e0307153. [PMID: 39509402 PMCID: PMC11542830 DOI: 10.1371/journal.pone.0307153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/01/2024] [Indexed: 11/15/2024] Open
Abstract
Microtubule-targeting agents are an important class of anti-cancer drugs; their full potential is however not realized because of significant myelotoxicity and neurotoxicity. We here report 3-nitropyridine analogues as a novel group of microtubule-targeting agents with potent anti-cancer effects against a broad range of cancer types. We show that these 3-nitropyridines induce cell cycle arrest in the G2-M phase and inhibit tubulin polymerization by interacting with tubulin. Determination of the tubulin-4AZA2996 structure by X-ray crystallography demonstrated that this class of compounds binds to the colchicine-site of tubulin. Furthermore, the anti-cancer effect was demonstrated both in vitro and in vivo in a murine heterotopic xenograft model of colon cancer. When administered intravenously, 4AZA2891 effectively inhibited cancer growth. Whereas 3-nitropyridine compounds do not induce myelotoxicity at pharmacological doses, the neurotoxicity associated with microtubule-targeting agents is still present.
Collapse
Affiliation(s)
- Jean Herman
- 4AZA Biosciences, Leuven, Belgium
- Interface Valorisation Platform (IVAP), KU Leuven, Leuven, Belgium
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
- Department of Pediatric Nephrology and Solid Organ Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Els Vanstreels
- Molecular Genetics and Therapeutics in Virology and Oncology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | | | - Andrea E. Prota
- Laboratory of Biomolecular Research, Center for Life Sciences, Paul Scherrer Institut, Würenlingen, Switzerland
| | - Natacha Gaillard
- Laboratory of Biomolecular Research, Center for Life Sciences, Paul Scherrer Institut, Würenlingen, Switzerland
| | - Ling-Jie Gao
- 4AZA Biosciences, Leuven, Belgium
- Interface Valorisation Platform (IVAP), KU Leuven, Leuven, Belgium
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical and Pharmacological Sciences, Rega Institute, KU Leuven, Leuven, Belgium
| | - Thomas Vercruysse
- Molecular Genetics and Therapeutics in Virology and Oncology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Leentje Persoons
- Molecular Genetics and Therapeutics in Virology and Oncology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | | | - Mark Waer
- 4AZA Biosciences, Leuven, Belgium
- Interface Valorisation Platform (IVAP), KU Leuven, Leuven, Belgium
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Piet Herdewijn
- 4AZA Biosciences, Leuven, Belgium
- Interface Valorisation Platform (IVAP), KU Leuven, Leuven, Belgium
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical and Pharmacological Sciences, Rega Institute, KU Leuven, Leuven, Belgium
| | - Thierry Louat
- 4AZA Biosciences, Leuven, Belgium
- Interface Valorisation Platform (IVAP), KU Leuven, Leuven, Belgium
| | - Michel O. Steinmetz
- Laboratory of Biomolecular Research, Center for Life Sciences, Paul Scherrer Institut, Würenlingen, Switzerland
- University of Basel, Basel, Switzerland
| | - Steven De Jonghe
- 4AZA Biosciences, Leuven, Belgium
- Interface Valorisation Platform (IVAP), KU Leuven, Leuven, Belgium
- Molecular Structural and Translational Virology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Ben Sprangers
- Interface Valorisation Platform (IVAP), KU Leuven, Leuven, Belgium
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
- Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Dirk Daelemans
- Molecular Genetics and Therapeutics in Virology and Oncology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Shi C, Yang B, He Z, Yang J, Li L, Song J, Xu S, Song W, Yang J. Discovery of novel 2-substituted 2, 3-dihydroquinazolin-4(1H)-one derivatives as tubulin polymerization inhibitors for anticancer therapy: The in vitro and in vivo biological evaluation. Eur J Med Chem 2024; 277:116766. [PMID: 39163776 DOI: 10.1016/j.ejmech.2024.116766] [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: 05/20/2024] [Revised: 07/22/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
A series of novel 2-substituted 2, 3-dihydroquinazolin-4(1H)-one derivatives were designed, synthesized and estimated for their in vitro antiproliferative activities against HepG2, U251, PANC-1, A549 and A375 cell lines. Among them, compound 32 was the most promising candidate, and displayed strong broad-spectrum anticancer activity. The mechanism studies revealed that compound 32 inhibited tubulin polymerization in vitro, disrupted cell microtubule networks, arrested the cell cycle at G2/M phase, and induced apoptosis by up-regulating the expression of cleaved PARP-1 and caspase-3. Furthermore, molecular docking analysis suggested that compound 32 well occupied the binding site of tubulin. In addition, compound 32 exhibited no significant activity against 30 different kinases respectively, indicating considerable selectivity. Moreover, compound 32 significantly inhibited the tumour growth of the HepG2 xenograft in a nude mouse model by oral gavage without apparent toxicity. These results demonstrated that some 2-substituted 2, 3- dihydroquinazolin-4(1H)-one derivatives bearing phenyl, biphenyl, naphthyl or indolyl side chain at C2-position might be potentially novel antitumor agents as tubulin polymerization inhibitors.
Collapse
Affiliation(s)
- Cai Shi
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Boning Yang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Institute of Pharmaceutical Process, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China; Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Zhaolong He
- Institute of Pharmaceutical Process, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Jingxiang Yang
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China
| | - Ling Li
- Institute of Pharmaceutical Process, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Jian Song
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shiqiang Xu
- Institute of Pharmaceutical Process, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China.
| | - Wei Song
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jian Yang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
33
|
Xu C, Wu C, Li L, Zhao H, Liu J, Peng X, Wang Y, Chen J. Discovery of novel thiophene[3,2-d]pyrimidine-based tubulin inhibitors with enhanced antitumor efficacy for combined use with anti-pd-l1 immunotherapy in melanoma. Eur J Med Chem 2024; 277:116791. [PMID: 39197251 DOI: 10.1016/j.ejmech.2024.116791] [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: 06/28/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024]
Abstract
Herein, we designed and synthesized a series of novel 2-methylthieno [3,2-d]pyrimidine analogues as tubulin inhibitors with antiproliferative activities at low nanomolar levels. Among them, compound DPP-21 displayed the most potent anti-proliferative activity against six cancer cell lines with an average IC50 of ∼6.23 nM, better than that of colchicine (IC50 = 9.26 nM). DPP-21 exerted its anti-cancer activity by suppressing the polymerization of tubulin with an IC50 of 2.4 μM. Furthermore, the crystal structure of DPP-21 in complex with tubulin was solved by X-ray crystallography to 2.94 Å resolution, confirming the direct binding of DPP-21 to the colchicine site. Moreover, DPP-21 arrested the cell cycle in the G2/M phase of mitosis, subsequently inducing tumor cell apoptosis. Additionally, DPP-21 was able to effectively inhibit the migration of cancer cells. Besides, DPP-21 exhibited significant in vivo anti-tumor efficacy in a B16-F10 melanoma tumor model with a TGI of 63.3 % (7 mg/kg) by intraperitoneal (i.p.) injection. Notably, the combination of DPP-21 with NP-19 (a PD-L1-targeting small molecule inhibitor reported by our group before) demonstrated enhanced anti-cancer efficacy in vivo. These results suggest that DPP-21 is a promising lead compound deserving further investigation as a potential anti-cancer agent.
Collapse
Affiliation(s)
- Chenglong Xu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Chengyong Wu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ling Li
- The Eighth Affiliated Hospital Sun Yat-sen University, 3025 Shennan Middle Road, Shenzhen, 518000, China
| | - Huiting Zhao
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Jin Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, 570228, China
| | - Xiaopeng Peng
- College of Pharmacy, Gannan Medical University, Ganzhou, 314000, China
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
34
|
Verghese D, Hamal S, Ghanem A, Kinninger A, Javier D, Ichikawa K, Benzing T, Krishnan S, Kianoush S, Hamidi H, Bagheri M, Abraham D, Deljavanghodrati M, Ghoto A, Aldana-Bitar J, Budoff M. Effect of colchicine on progression of known coronary atherosclerosis in patients with STable CoROnary artery disease CoMpared to placebo (EKSTROM) trial-rationale and design. Am Heart J 2024; 277:20-26. [PMID: 39029568 DOI: 10.1016/j.ahj.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/17/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Cardiovascular disease is the major cause of mortality in the United States. Despite lifestyle modification and traditional risk factor control residual inflammatory risk remains an untreated concern. Colchicine is an oral, medication that has been used for gout, mediterranean fever and pericarditis for decades. In recent trials, colchicine has been shown to reduce major adverse cardiovascular events, however the mechanism of benefit remains unclear. The objective of the randomized, double-blind, placebo controlled EKSTROM trial is to evaluate the effects of colchicine 0.5mg/day on atherosclerotic plaque. METHODS Eighty-four participants will be enrolled after obtaining informed consent and followed for 12 months. Eligible patients will be randomly assigned to colchicine 0.5mg/day or placebo in a 1:1 fashion as add-on to their standard of care. All participants will undergo coronary computed tomography angiography (CCTA) at baseline and at 12 months. RESULTS As of November 2023, the study is 100% enrolled with an expected end of study by the second quarter of 2024. The primary endpoint is change in low attenuation plaque volume as measured by CCTA. Secondary endpoints include change in volume of different plaque types (including total atheroma volume, noncalcified plaque volume, dense calcified plaque volume, remodeling index), change in inflammatory markers (IL-6, IL-1β, IL-18, hs-CRP), change in pericoronary adipose tissue attenuation, change in epicardial adipose tissue volume and attenuation and change in brachial flow mediated dilation. CONCLUSION EKSTROM is the first randomized study to assess the effects of colchicine on plaque progression, pericoronary and epicardial fat. EKSTROM will provide important information on the mechanistic effects of colchicine on the cardiovascular system. TRIAL REGISTRATION Registry: clinicaltrials.gov, Registration Number: NCT06342609 url: https://www. CLINICALTRIALS gov/study/NCT06342609?term=EKSTROM&rank=1.
Collapse
Affiliation(s)
- Dhiran Verghese
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California; Department of Medicine, Division of Cardiovascular Medicine, NCH Rooney Heart Institute, Naples, Florida.
| | - Sajad Hamal
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Ahmed Ghanem
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - April Kinninger
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Denise Javier
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Keshi Ichikawa
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Travis Benzing
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Srikanth Krishnan
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Sina Kianoush
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Hossein Hamidi
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Marziyeh Bagheri
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Divya Abraham
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Mina Deljavanghodrati
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Ayesha Ghoto
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Jairo Aldana-Bitar
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| | - Matthew Budoff
- Department of Medicine, Division of Cardiovascular Medicine, The Lundquist Institute, Harbor UCLA Medical Center, Torrance, California
| |
Collapse
|
35
|
Anisimov MN, Boichenko MA, Shorokhov VV, Borzunova JN, Janibekova M, Mustyatsa VV, Lifshits IA, Plodukhin AY, Andreev IA, Ratmanova NK, Zhokhov SS, Tarasenko EA, Ipatova DA, Pisarev AR, Vorobjev IA, Trushkov IV, Ivanova OA, Gudimchuk NB. Synthesis and evaluation of tetrahydropyrrolo[1,2- a]quinolin-1(2 H)-ones as new tubulin polymerization inhibitors. RSC Med Chem 2024; 16:d4md00541d. [PMID: 39464648 PMCID: PMC11499956 DOI: 10.1039/d4md00541d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024] Open
Abstract
Here we explored new 1,5-disubstituted pyrrolidin-2-ones 1, 2 and 5-aryl-3,3a,4,5-tetrahydropyrrolo[1,2-a]quinoline-1(2H)-ones 3 as inhibitors of tubulin polymerization. We evaluated their effects on microtubule dynamics in vitro and on the proliferation of A549 cells, using flow cytometry-based cell cycle analysis. The results were verified with phase-contrast microscopy in three cancer cell lines: A549, HeLa and MCF-7. Guided by molecular modeling of the interactions between tubulin and the most active of the identified compounds, we designed, synthesized, and tested the 3-hydroxyphenyl-substituted compound 3c. This compound was further shown to bind to the colchicine site of tubulin and reduce microtubule growth rates in vitro. Moreover, compound 3c arrested division of the A549 cells in the low micromolar range (IC50 = 5.9 μM) and exhibited cytotoxicity against four different cell lines in the MTT assay for cell proliferation. Our findings demonstrate that 5-aryltetrahydropyrrolo[1,2-a]quinoline-1(2H)-one is a promising scaffold for the development of novel tubulin polymerization inhibitors.
Collapse
Affiliation(s)
- Mikhail N Anisimov
- Department of Physics, M.V. Lomonosov Moscow State University Moscow 119991 Russia
- Center for theoretical problems of physicochemical pharmacology Moscow 109029 Russia
| | - Maksim A Boichenko
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Vitaly V Shorokhov
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Julia N Borzunova
- Department of Physics, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | | | - Vadim V Mustyatsa
- Center for theoretical problems of physicochemical pharmacology Moscow 109029 Russia
- National Laboratory Astana Astana 010000 Kazakhstan
| | - Ilya A Lifshits
- Department of Physics, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Andrey Yu Plodukhin
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Ivan A Andreev
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow 117997 Russia
| | - Nina K Ratmanova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow 117997 Russia
| | - Sergey S Zhokhov
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Elena A Tarasenko
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Daria A Ipatova
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Alexander R Pisarev
- Faculty of Biology and Biotechnologies, Higher School of Economics Moscow 117418 Russia
| | - Ivan A Vorobjev
- National Laboratory Astana Astana 010000 Kazakhstan
- Department of Biology, School of Sciences and Humanities, Nazarbayev University Astana 010000 Kazakhstan
- Department of Biology, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Igor V Trushkov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Moscow 119991 Russia
| | - Olga A Ivanova
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Nikita B Gudimchuk
- Department of Physics, M.V. Lomonosov Moscow State University Moscow 119991 Russia
- Center for theoretical problems of physicochemical pharmacology Moscow 109029 Russia
| |
Collapse
|
36
|
Wu C, Zhang L, Zhou Z, Tan L, Wang Z, Guo C, Wang Y. Discovery and mechanistic insights into thieno[3,2-d]pyrimidine and heterocyclic fused pyrimidines inhibitors targeting tubulin for cancer therapy. Eur J Med Chem 2024; 276:116649. [PMID: 38972078 DOI: 10.1016/j.ejmech.2024.116649] [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: 04/16/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Guided by the X-ray cocrystal structure of the lead compound 4a, we developed a series of thieno[3,2-d]pyrimidine and heterocyclic fused pyrimidines demonstrating potent antiproliferative activity against four tumor cell lines. Two analogs, 13 and 25d, exhibited IC50 values around 1 nM and overcame P-glycoprotein (P-gp)-mediated multidrug resistance (MDR). At low concentrations, 13 and 25d inhibited both the colony formation of SKOV3 cells in vitro and tubulin polymerization. Furthermore, mechanistic studies showed that 13 and 25d induced G2/M phase arrest and apoptosis in SKOV3 cells, as well as dose-dependent inhibition of tumor cell migration and invasion at low concentrations. Most notably, the X-ray cocrystal structures of compounds 4a, 25a, and the optimal molecule 13 in complex with tubulin were elucidated. This study identifies thieno[3,2-d]pyrimidine and heterocyclic fused pyrimidines as representatives of colchicine-binding site inhibitors (CBSIs) with potent antiproliferative activity.
Collapse
Affiliation(s)
- Chengyong Wu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lele Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhilan Zhou
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lun Tan
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhijia Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cuiyu Guo
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
37
|
Lu B, Xu L, Chen H, Yu G, Khow K, Yang S, Dinker A, Njoo E. C-4 analogues of podophyllotoxin as tubulin inhibitors: synthesis, biological evaluation, and structure-activity relationship. Nat Prod Res 2024:1-7. [PMID: 39360483 DOI: 10.1080/14786419.2024.2410410] [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: 02/13/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
The diversity of lignan small molecules derived from podophyllotoxin, a non-covalent tubulin inhibitor isolated from the Podophyllum family, has led to the clinical development of FDA-approved anticancer agents etoposide and teniposide. While these two compounds share the same tetracyclic core as podophyllotoxin, two subtle structural changes-4' demethylation on the aromatic ring and stereospecific glycosylation at the C-4 hydroxyl-result in an alternate biological mechanism. Given the immense pharmacological importance of altering the C-4 position, we synthesised and evaluated a systematic library of diversified esters to establish a structure-activity relationship regarding modification at C-4 on the properties of podophyllotoxin. We determined the biological activity of these esters through cell viability assays, computer docking models, tubulin polymerisation assays, and cell cycle analysis. Altogether, we demonstrate that increasing steric hindrance at C-4 leads to a loss in potency against human cancer cells but has a significantly lesser impact on cell-free tubulin inhibition.
Collapse
Affiliation(s)
- Breanna Lu
- Department of Chemistry, Biochemistry & Physics, Aspiring Scholars Directed Research Program (ASDRP), Fremont, CA, USA
| | - Lexi Xu
- Department of Chemistry, Biochemistry & Physics, Aspiring Scholars Directed Research Program (ASDRP), Fremont, CA, USA
| | - Harriet Chen
- Department of Chemistry, Biochemistry & Physics, Aspiring Scholars Directed Research Program (ASDRP), Fremont, CA, USA
| | - Grace Yu
- Department of Chemistry, Biochemistry & Physics, Aspiring Scholars Directed Research Program (ASDRP), Fremont, CA, USA
| | - Kimberly Khow
- Department of Chemistry, Biochemistry & Physics, Aspiring Scholars Directed Research Program (ASDRP), Fremont, CA, USA
| | - Stella Yang
- Department of Chemistry, Biochemistry & Physics, Aspiring Scholars Directed Research Program (ASDRP), Fremont, CA, USA
| | - Arushi Dinker
- Department of Chemistry, Biochemistry & Physics, Aspiring Scholars Directed Research Program (ASDRP), Fremont, CA, USA
| | - Edward Njoo
- Department of Chemistry, Biochemistry & Physics, Aspiring Scholars Directed Research Program (ASDRP), Fremont, CA, USA
| |
Collapse
|
38
|
Tsai JF, Yu FY, Liu BH. Citrinin disrupts microtubule assembly in cardiac cells: Impact on mitochondrial organization and function. CHEMOSPHERE 2024; 365:143352. [PMID: 39293683 DOI: 10.1016/j.chemosphere.2024.143352] [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: 05/20/2024] [Revised: 08/31/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
Citrinin (CTN) is a mycotoxin commonly present in various foods and feeds worldwide, as well as dietary supplements in Asian countries, but the risks and cellular mechanisms associated with its cardiotoxicity remains unclear. In this study, RNA-seq analysis of CTN-treated H9c2 cardiac cells demonstrated significant perturbations in pathways related to microtubule cytoskeleton and mitochondrial network organization. CTN disrupted microtubule polymerization and downregulated mRNA levels of microtubule-assembling genes, Map2 and Tpx2, in H9c2 cardiac cells. Additionally, CTN interfered with the distribution of mitochondrial network along the microtubules, leading to the accumulation of dysfunctional mitochondria characterized by elevated superoxide levels and reduced membrane potential. This disruption also caused the buildup of lysosomes and ubiquitinated proteins, which hindered waste clearance in microtubule-disassembled H9c2 cells. Molecular docking analysis indicated that CTN could bind to the colchicine binding site on β-tubulin, thereby mimicking the microtubule-disrupting effect of colchicine. This study provides morphological, transcriptomic, and mechanistic evidence to elucidate the cardiotoxic mechanisms of CTN, which involve the dysregulated microtubule network, subsequent mitochondrial mislocalization, and impaired proteolysis of damaged proteins/organelles in cardiac cells. Our findings may enhance the fundamental understanding and facilitate future risk assessment of CTN.
Collapse
Affiliation(s)
- Jui-Feng Tsai
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Feng-Yih Yu
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Biing-Hui Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
39
|
Aljuhani A, Nafie MS, Albujuq NR, Hourani W, Albelwi FF, Darwish KM, Samir Ayed A, Reda Aouad M, Rezki N. Unveiling the anti-cancer potentiality of phthalimide-based Analogues targeting tubulin polymerization in MCF-7 cancerous Cells: Rational design, chemical Synthesis, and Biological-coupled Computational investigation. Bioorg Chem 2024; 153:107827. [PMID: 39321715 DOI: 10.1016/j.bioorg.2024.107827] [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: 07/06/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
The present study deals with an anti-cancer investigation of an array of phthalimide-1,2,3-triazole molecular conjugates with various sulfonamide fragments against human breast MCF-7 and prostate PC3 cancer cell lines. The targeted 1,2,3-triazole derivatives 4a-l and 6a-c were synthesized from focused phthalimide-based alkyne precursors using a facile click synthesis approach and were thoroughly characterized using several spectroscopic techniques (IR, 1H, 13C NMR, and elemental analysis). The hybrid click adducts 4b, 4 h, and 6c displayed cytotoxic potency (IC50 values of 1.49, 1.07, and 0.56 μM, respectively) against MCF-7 cells. On the contrary, none of the synthesized compounds showed apparent cytotoxic efficacy for PC3 cells (IC50 ranging from 9.87- >100 μM). As a part of the mechanism analysis, compound 6c demonstrated a potent inhibitory effect (78.3 % inhibition) of tubulin polymerization in vitro with an IC50 value of 6.53 µM. In addition, biological assays showed that compound 6c could prompt apoptotic cell death and induce G2/M cell cycle arrest in MCF-7 cells. Accordingly, compound 6c can be further developed as an anti-breast cancer agent through apoptosis-induction.
Collapse
Affiliation(s)
- Ateyatallah Aljuhani
- Department of Chemistry, Faculty of Science, Taibah University, Al-Madinah Al-Munawarah 41477, Saudi Arabia.
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah P.O. 27272, United Arab Emirates (UAE); Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, P.O. 41522, Egypt.
| | - Nader R Albujuq
- Department of Chemistry, School of Science, The University of Jordan, Amman 11942, Jordan.
| | - Wafa Hourani
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Philadelphia University, Amman 19392, Jordan.
| | - Fawzia F Albelwi
- Department of Chemistry, Faculty of Science, Taibah University, Al-Madinah Al-Munawarah 41477, Saudi Arabia.
| | - Khaled M Darwish
- Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt.
| | - Aya Samir Ayed
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, P.O. 41522, Egypt.
| | - Mohamed Reda Aouad
- Department of Chemistry, Faculty of Science, Taibah University, Al-Madinah Al-Munawarah 41477, Saudi Arabia.
| | - Nadjet Rezki
- Department of Chemistry, Faculty of Science, Taibah University, Al-Madinah Al-Munawarah 41477, Saudi Arabia.
| |
Collapse
|
40
|
Thornburg-Suresh EJC, Summers DW. Microtubules, Membranes, and Movement: New Roles for Stathmin-2 in Axon Integrity. J Neurosci Res 2024; 102:e25382. [PMID: 39253877 PMCID: PMC11407747 DOI: 10.1002/jnr.25382] [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: 05/07/2024] [Revised: 08/06/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024]
Abstract
Neurons establish functional connections responsible for how we perceive and react to the world around us. Communication from a neuron to its target cell occurs through a long projection called an axon. Axon distances can exceed 1 m in length in humans and require a dynamic microtubule cytoskeleton for growth during development and maintenance in adulthood. Stathmins are microtubule-associated proteins that function as relays between kinase signaling and microtubule polymerization. In this review, we describe the prolific role of Stathmins in microtubule homeostasis with an emphasis on emerging roles for Stathmin-2 (Stmn2) in axon integrity and neurodegeneration. Stmn2 levels are altered in Amyotrophic Lateral Sclerosis and loss of Stmn2 provokes motor and sensory neuropathies. There is growing potential for employing Stmn2 as a disease biomarker or even a therapeutic target. Meeting this potential requires a mechanistic understanding of emerging complexity in Stmn2 function. In particular, Stmn2 palmitoylation has a surprising contribution to axon maintenance through undefined mechanisms linking membrane association, tubulin interaction, and axon transport. Exploring these connections will reveal new insight on neuronal cell biology and novel opportunities for disease intervention.
Collapse
Affiliation(s)
| | - Daniel W Summers
- Department of Biology, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
41
|
Yan W, Zhou Y, Yuan X, Bai P, Tang M, Chen L, Wei H, Yang J. The cytotoxic natural compound erianin binds to colchicine site of β-tubulin and overcomes taxane resistance. Bioorg Chem 2024; 150:107569. [PMID: 38905886 DOI: 10.1016/j.bioorg.2024.107569] [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: 03/26/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
Erianin, a natural compound derived from Dendrobium, has shown significant anticancer properties against a wide range of cancer cells. Despite the identification of multiple mechanisms of action for erianin, none of these mechanisms fully account for its broad-spectrum effect. In this study, we aimed to identify the cellular target and underlying mechanism responsible for the broad-spectrum antitumor effects of erianin. We found that erianin effectively inhibited tubulin polymerization in cancer cells and purified tubulin. Through competition binding assays and X-ray crystallography, it was revealed that erianin bound to the colchicine site of β-tubulin. Importantly, the X-ray crystal structure of the tubulin-erianin complex was solved, providing clear insight into the orientation and position of erianin in the colchicine-binding site. Erianin showed activity against paclitaxel-resistant cells, evidenced by G2/M cell cycle arrest, apoptosis-related PARP and Caspase-3 cleavage, and in vivo xenograft studies. The study concluded that erianin bound reversibly to the colchicine site of β-tubulin, inhibited tubulin polymerization, and displayed anticancer activity against paclitaxel-resistant cells, offering valuable insights for further exploration as potential anticancer agents.
Collapse
Affiliation(s)
- Wei Yan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yongzhao Zhou
- Integrated Care Management Center, West China Hospital, Sichuan University, China.
| | - Xue Yuan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Peng Bai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lijuan Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haoche Wei
- Department of General Surgery, Gastric Cancer Center, Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Jianhong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
42
|
Abd-Rabo ZS, Serry AM, George RF. An overview of pyridazin-3(2 H)-one: a core for developing bioactive agents targeting cardiovascular diseases and cancer. Future Med Chem 2024; 16:1685-1703. [PMID: 39105606 PMCID: PMC11370926 DOI: 10.1080/17568919.2024.2379234] [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: 02/28/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024] Open
Abstract
Cardiovascular diseases (CVDs) and cancer are the top two leading causes of death globally. Vasodilators are commonly used to treat various CVDs. In cancer treatment, targeted anticancer agents have been developed to minimize side effects compared with traditional chemotherapy. Many hypertension patients are more prone to cancer, a case known as reverse cardio-oncology. This leads to the search for drugs with dual activity or repurposing strategy to discover new therapeutic uses for known drugs. Recently, medicinal chemists have shown great interest in synthesizing pyridazinone derivatives due to their significant biological activities in tackling these critical health challenges. This review will concentrate on pyridazin-3(2H)-one-containing compounds as vasodilators and anticancer agents, along with a brief overview of various methods for their synthesis.
Collapse
Affiliation(s)
- Zeinab S Abd-Rabo
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology & Information MTI, Cairo, 11571, Egypt
| | - Aya M Serry
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology & Information MTI, Cairo, 11571, Egypt
| | - Riham F George
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
43
|
Serena NN, Besati M, Nadia NAC, Yaghoobi M, Cédric Y, Ciancia C, Sidiki NNA, Payne VK, Mbida M, Hu H. In Vitro and In Silico Anthelmintic Activity of Extracts of Lannea kerstingii and Ficus thonningii on Heligmosomoides polygyrus. J Parasitol Res 2024; 2024:1858154. [PMID: 39131749 PMCID: PMC11316912 DOI: 10.1155/2024/1858154] [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: 01/25/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Background: The aim of this study was to assess the anthelmintic activity of Lannea kerstingii and Ficus thonningii, on a nematode model, to promote their use in the Cameroonian pharmacopoeia for the treatment of helminthiases. Methods: One nematode was used, Heligmosomoides polygyrus. First, the effect of the extracts on the eggs and larval stages (L1, L2, and L3) of H. polygyrus was evaluated, 100 μL of extract and 100 μL of parasite suspension (containing 50 eggs) were mixed in a 96-well microplate. The 96-well microplate was incubated for 20 h at 25°C in the WMicroTracker which measures the motility of the worms at various concentrations. Finally, docking studies were conducted by using the Glide module in Schrodinger Maestro. Results: The ethanolic extract of L. kerstingii with the half maximal inhibitory concentration (IC50) of 0.1371 mg/mL produced a higher ovicidal effect than the effect produced by other extracts of these plants. However, with an IC50 of 0.31 mg/mL, the aqueous extract of F. thonningii showed the greatest effect on the L2 stage. The aqueous and ethanolic extracts of L. kerstingii and F. thonningii inhibited the development of the L3 larvae of H. polygyrus with a better effect for the ethanolic extracts. Conclusion: The use of L. kerstingii and F. thonningii for the treatment of helminthiasis has been proved in vitro and in silico by this research. However, more research is required, especially on the acute toxicity and in vivo anthelmintic efficacy to validate this scientific investigation.
Collapse
Affiliation(s)
- Ndjinkeu Ntcheuzing Serena
- Department of Animal BiologyFaculty of ScienceUniversity of Dschang, P.O. Box 067, Dschang, Cameroon
- Laboratory of Tropical and Emerging Infectious Diseases, Dschang, Cameroon
| | - Masoud Besati
- Institute for Integrative Systems Biology (I2SysBio)CSIC−University of Valencia 46980, Paterna, Spain
| | - Noumedem Anangmo Christelle Nadia
- Laboratory of Tropical and Emerging Infectious Diseases, Dschang, Cameroon
- Department of MicrobiologyHaematology and Immunology Faculty of Medicine and Pharmaceutical SciencesUniversity of Dschang, P.O. Box 96, Dschang, Cameroon
| | - Mahdi Yaghoobi
- Molecular Design and SynthesisDepartment of ChemistryKU Leuven, Celestijnenlaan 200F B-3001, Leuven, Belgium
| | - Yamssi Cédric
- Laboratory of Tropical and Emerging Infectious Diseases, Dschang, Cameroon
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of Bamenda, P.O. Box 39, Bambili, Cameroon
| | - Claire Ciancia
- Wellcome Centre for Molecular ParasitologySchool for Infection and ImmunityUniversity of Glasgow, Glasgow, UK
| | - Ngouyamsa Nsapkain Aboubakar Sidiki
- Department of Animal BiologyFaculty of ScienceUniversity of Dschang, P.O. Box 067, Dschang, Cameroon
- Laboratory of Tropical and Emerging Infectious Diseases, Dschang, Cameroon
| | - Vincent Khan Payne
- Department of Animal BiologyFaculty of ScienceUniversity of Dschang, P.O. Box 067, Dschang, Cameroon
- Laboratory of Tropical and Emerging Infectious Diseases, Dschang, Cameroon
| | - Mpoame Mbida
- Department of Animal BiologyFaculty of ScienceUniversity of Dschang, P.O. Box 067, Dschang, Cameroon
| | - Haibo Hu
- Laboratory of Tropical and Emerging Infectious Diseases, Dschang, Cameroon
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of Bamenda, P.O. Box 39, Bambili, Cameroon
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese MedicineNational Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources BranchSchool of PharmacyGannan Medical University, Ganzhou 341000, China
| |
Collapse
|
44
|
Peng Y, Zhang Y, Fang R, Jiang H, Lan G, Xu Z, Liu Y, Nie Z, Ren L, Wang F, Zhang S, Ma Y, Yang P, Ge H, Zhang W, Luo C, Li A, He W. Target Identification and Mechanistic Characterization of Indole Terpenoid Mimics: Proper Spindle Microtubule Assembly Is Essential for Cdh1-Mediated Proteolysis of CENP-A. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305593. [PMID: 38873820 PMCID: PMC11304278 DOI: 10.1002/advs.202305593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 04/23/2024] [Indexed: 06/15/2024]
Abstract
Centromere protein A (CENP-A), a centromere-specific histone H3 variant, is crucial for kinetochore positioning and chromosome segregation. However, its regulatory mechanism in human cells remains incompletely understood. A structure-activity relationship (SAR) study of the cell-cycle-arresting indole terpenoid mimic JP18 leads to the discovery of two more potent analogs, (+)-6-Br-JP18 and (+)-6-Cl-JP18. Tubulin is identified as a potential cellular target of these halogenated analogs by using the drug affinity responsive target stability (DARTS) based method. X-ray crystallography analysis reveals that both molecules bind to the colchicine-binding site of β-tubulin. Treatment of human cells with microtubule-targeting agents (MTAs), including these two compounds, results in CENP-A accumulation by destabilizing Cdh1, a co-activator of the anaphase-promoting complex/cyclosome (APC/C) E3 ubiquitin ligase. This study establishes a link between microtubule dynamics and CENP-A accumulation using small-molecule tools and highlights the role of Cdh1 in CENP-A proteolysis.
Collapse
Affiliation(s)
- Yan Peng
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Yumeng Zhang
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Ruan Fang
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
- State Key Laboratory of Chemical BiologyShanghai Institute of Organic ChemistryUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200032China
| | - Hao Jiang
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Gongcai Lan
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Zhou Xu
- State Key Laboratory of Chemical BiologyShanghai Institute of Organic ChemistryUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200032China
| | - Yajie Liu
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Zhaoyang Nie
- State Key Laboratory of Chemical BiologyShanghai Institute of Organic ChemistryUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200032China
- Henan Institute of Advanced Technology and College of ChemistryZhengzhou UniversityZhengzhou450001China
| | - Lu Ren
- State Key Laboratory of Chemical BiologyShanghai Institute of Organic ChemistryUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200032China
| | - Fengcan Wang
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Shou‐De Zhang
- State Key Laboratory of Plateau Ecology and AgricultureQinghai UniversityXining810016China
| | - Yuyong Ma
- State Key Laboratory of Chemical BiologyShanghai Institute of Organic ChemistryUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200032China
| | - Peng Yang
- State Key Laboratory of Chemical BiologyShanghai Institute of Organic ChemistryUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200032China
- Henan Institute of Advanced Technology and College of ChemistryZhengzhou UniversityZhengzhou450001China
| | - Hong‐Hua Ge
- Institute of Physical Science and Information TechnologyAnhui UniversityHefei230601China
| | - Wei‐Dong Zhang
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Cheng Luo
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Ang Li
- State Key Laboratory of Chemical BiologyShanghai Institute of Organic ChemistryUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200032China
- Henan Institute of Advanced Technology and College of ChemistryZhengzhou UniversityZhengzhou450001China
| | - Weiwei He
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| |
Collapse
|
45
|
Steinmetz MO, Prota AE. Structure-based discovery and rational design of microtubule-targeting agents. Curr Opin Struct Biol 2024; 87:102845. [PMID: 38805950 DOI: 10.1016/j.sbi.2024.102845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
Microtubule-targeting agents (MTAs) have demonstrated remarkable efficacy as antitumor, antifungal, antiparasitic, and herbicidal agents, finding applications in the clinical, veterinary, and agrochemical industry. Recent advances in tubulin and microtubule structural biology have provided powerful tools that pave the way for the rational design of innovative small-molecule MTAs for future basic and applied life science applications. In this mini-review, we present the current status of the tubulin and microtubule structural biology field, the recent impact it had on the discovery and rational design of MTAs, and exciting avenues for future MTA research.
Collapse
Affiliation(s)
- Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland; University of Basel, Biozentrum, 4056 Basel, Switzerland.
| | - Andrea E Prota
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland.
| |
Collapse
|
46
|
Nagavath R, Thupurani MK, Badithapuram V, Manchal R, Vasam CS, Thirukovela NS. Organo NHC catalyzed aqueous synthesis of 4β-isoxazole-podophyllotoxins: in vitro anticancer, caspase activation, tubulin polymerization inhibition and molecular docking studies. RSC Adv 2024; 14:23574-23582. [PMID: 39070249 PMCID: PMC11276401 DOI: 10.1039/d4ra04297b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
We present, for the first time, the organo-N-heterocyclic carbene (NHC) catalyzed 1,3-dipolar cycloaddition of 4β-O-propargyl podophyllotoxin (1) with in situ aromatic nitrile oxides to afford regioselective 4β-isoxazolepodophyllotoxin hybrids (6a-n) in benign aqueous-organic media. Preliminary anticancer activity results showed that compound 6e displayed superior activity against MCF-7, HeLa and MIA PaCa2 human cell lines compared with podophyllotoxin. Compounds 6j and 6n showed greater activity against the MCF-7 cell line than the positive control. Caspase activation studies revealed that compound 6e at 20 μg ml-1 concentration had greater caspase 3/7 activation in MCF-7 and MIAPaCa2 cells than podophyllotoxin. Furthermore, in vitro tubulin polymerization inhibition studies revealed that compound 6e showed comparable activity with podophyllotoxin. Finally, in silico molecular docking studies of compounds 6e, 6j, 6n and podophyllotoxin on α,β-tubulin (pdb id 1SA0) revealed that compound 6n showed excellent binding energies and inhibition constants compared with podophyllotoxin.
Collapse
Affiliation(s)
- Rajkumar Nagavath
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | - Murali Krishna Thupurani
- Department of Biotechnology, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | - Vinitha Badithapuram
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | - Ravinder Manchal
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | | | - Narasimha Swamy Thirukovela
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| |
Collapse
|
47
|
Moussaoui M, Baammi S, Soufi H, Baassi M, El Allali A, Belghiti ME, Daoud R, Belaaouad S. QSAR, ADMET, molecular docking, and dynamics studies of 1,2,4-triazine-3(2H)-one derivatives as tubulin inhibitors for breast cancer therapy. Sci Rep 2024; 14:16418. [PMID: 39013949 PMCID: PMC11252338 DOI: 10.1038/s41598-024-66877-2] [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: 03/18/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
Breast cancer remains a leading cause of cancer-related deaths among women globally, necessitating the development of more effective therapeutic agents with minimal side effects. This study explores novel 1,2,4-triazine-3(2H)-one derivatives as potential inhibitors of Tubulin, a pivotal protein in cancer cell division, highlighting a targeted approach in cancer therapy. Using an integrated computational approach, we combined quantitative structure-activity relationship (QSAR) modeling, ADMET profiling, molecular docking, and molecular dynamics simulations to evaluate and predict the efficacy and stability of these compounds. Our QSAR models, developed through rigorous statistical analysis, revealed that descriptors such as absolute electronegativity and water solubility significantly influence inhibitory activity, achieving a predictive accuracy (R2) of 0.849. Molecular docking studies identified compounds with high binding affinities, particularly Pred28, which exhibited the best docking score of - 9.6 kcal/mol. Molecular dynamics simulations conducted over 100 ns provided further insights into the stability of these interactions. Pred28 demonstrated notable stability, with the lowest root mean square deviation (RMSD) of 0.29 nm and root mean square fluctuation (RMSF) values indicative of a tightly bound conformation to Tubulin. The novelty of this work lies in its methodological rigor and the integration of multiple advanced computational techniques to pinpoint compounds with promising therapeutic potential. Our findings advance the current understanding of Tubulin inhibitors and open avenues for the synthesis and experimental validation of these compounds, aiming to offer new solutions for breast cancer treatment.
Collapse
Affiliation(s)
- Mohamed Moussaoui
- Laboratory of Physical Chemistry of Materials, Faculty of Sciences Ben M'Sick, Hassan II University of Casablanca, Casablanca, Morocco.
| | - Soukayna Baammi
- Bioinformatics Laboratory, College of Computing, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Hatim Soufi
- Laboratory of Physical Chemistry of Materials, Faculty of Sciences Ben M'Sick, Hassan II University of Casablanca, Casablanca, Morocco
| | - Mouna Baassi
- Laboratory of Physical Chemistry of Materials, Faculty of Sciences Ben M'Sick, Hassan II University of Casablanca, Casablanca, Morocco
| | - Achraf El Allali
- Bioinformatics Laboratory, College of Computing, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - M E Belghiti
- Laboratory of Physical Chemistry of Materials, Faculty of Sciences Ben M'Sick, Hassan II University of Casablanca, Casablanca, Morocco
- Laboratory of Nernest Technology, 163 Willington Street, Sherbrook, QC, J1H5C7, Canada
| | - Rachid Daoud
- Chemical and Biochemical Sciences-Green Processing Engineering, Mohammed VI Polytechnic University, Ben Guerir, Morocco.
| | - Said Belaaouad
- Laboratory of Physical Chemistry of Materials, Faculty of Sciences Ben M'Sick, Hassan II University of Casablanca, Casablanca, Morocco
| |
Collapse
|
48
|
La Monica G, Bono A, Alamia F, Lauria A, Martorana A. Bioisosteric heterocyclic analogues of natural bioactive flavonoids by scaffold-hopping approaches: State-of-the-art and perspectives in medicinal chemistry. Bioorg Med Chem 2024; 109:117791. [PMID: 38870715 DOI: 10.1016/j.bmc.2024.117791] [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: 03/07/2024] [Revised: 05/13/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
The flavonoid family is a set of well-known bioactive natural molecules, with a wide range of potential therapeutic applications. Despite the promising results obtained in preliminary in vitro/vivo studies, their pharmacokinetic and pharmacodynamic profiles are severely compromised by chemical instability. To address this issue, the scaffold-hopping approach is a promising strategy for the structural optimization of natural leads to discover more potent analogues. In this scenario, this Perspective provides a critical analysis on how the replacement of the chromon-4-one flavonoid core with other bioisosteric nitrogen/sulphur heterocycles might affect the chemical, pharmaceutical and biological properties of the resulting new chemical entities. The investigated derivatives were classified on the basis of their biological activity and potential therapeutic indications. For each session, the target(s), the specific mechanism of action, if available, and the key pharmacophoric moieties were highlighted, as revealed by X-ray crystal structures and in silico structure-based studies. Biological activity data, in vitro/vivo studies, were examined: a particular focus was given on the improvements observed with the new heterocyclic analogues compared to the natural flavonoids. This overview of the scaffold-hopping advantages in flavonoid compounds is of great interest to the medicinal chemistry community to better exploit the vast potential of these natural molecules and to identify new bioactive molecules.
Collapse
Affiliation(s)
- Gabriele La Monica
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy
| | - Alessia Bono
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy
| | - Federica Alamia
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy
| | - Antonino Lauria
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy
| | - Annamaria Martorana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy.
| |
Collapse
|
49
|
Mariotto E, Canton M, Marchioro C, Brancale A, Hamel E, Varani K, Vincenzi F, De Ventura T, Padroni C, Viola G, Romagnoli R. Synthesis and Biological Evaluation of Novel 2-Aroyl Benzofuran-Based Hydroxamic Acids as Antimicrotubule Agents. Int J Mol Sci 2024; 25:7519. [PMID: 39062759 PMCID: PMC11277476 DOI: 10.3390/ijms25147519] [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: 06/11/2024] [Revised: 07/02/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Because of synergism between tubulin and HDAC inhibitors, we used the pharmacophore fusion strategy to generate potential tubulin-HDAC dual inhibitors. Drug design was based on the introduction of a N-hydroxyacrylamide or a N-hydroxypropiolamide at the 5-position of the 2-aroylbenzo[b]furan skeleton, to produce compounds 6a-i and 11a-h, respectively. Among the synthesized compounds, derivatives 6a, 6c, 6e, 6g, 11a, and 11c showed excellent antiproliferative activity, with IC50 values at single- or double-digit nanomolar levels, against the A549, HT-29, and MCF-7 cells resistant towards the control compound combretastatin A-4 (CA-4). Compounds 11a and 6g were also 10-fold more active than CA-4 against the Hela cell line. When comparing the inhibition of tubulin polymerization versus the HDAC6 inhibitory activity, we found that 6a-g, 6i, 11a, 11c, and 11e, although very potent as inhibitors of tubulin assembly, did not have significant inhibitory activity against HDAC6.
Collapse
Affiliation(s)
- Elena Mariotto
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35128 Padova, Italy; (E.M.); (M.C.); (C.M.); (G.V.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Martina Canton
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35128 Padova, Italy; (E.M.); (M.C.); (C.M.); (G.V.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Chiara Marchioro
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35128 Padova, Italy; (E.M.); (M.C.); (C.M.); (G.V.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Andrea Brancale
- Department of Organic Chemistry, University of Chemistry and Technology, Prague, 166 28 Prague, Czech Republic;
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA;
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (K.V.); (F.V.)
| | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (K.V.); (F.V.)
| | - Tiziano De Ventura
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Chiara Padroni
- Medicinal Chemistry Department, Integrated Drug Discovery, Aptuit, an Evotec Company, 37135 Verona, Italy;
| | - Giampietro Viola
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35128 Padova, Italy; (E.M.); (M.C.); (C.M.); (G.V.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Romeo Romagnoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| |
Collapse
|
50
|
Khairuzzaman M, Hasan MM, Ali MT, Mamun AA, Akter S, Nasrin P, Islam MK, Nahar AU, Sarker DK, Hamdi OAA, Uddin SJ, Seidel V, Shilpi JA. Anthelmintic screening of Bangladeshi medicinal plants and related phytochemicals using in vitro and in silico methods: An ethnobotanical perspective. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118132. [PMID: 38565411 DOI: 10.1016/j.jep.2024.118132] [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: 12/18/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Infections caused by parasitic worms or helminth continue to pose a great burden on human and animal health, particularly in underdeveloped tropical and subtropical countries where they are endemic. Current anthelmintic drugs present serious limitations and the emergence of drug resistance has made it increasingly challenging to combat such infections (helminthiases). In Bangladesh, medicinal plants are often used by indigenous communities for the treatment of helminthiases. Knowledge on such plants along with screening for their anthelmintic activity has the potential to lead to the discovery of phytochemicals that could serve as novel molecular scaffolds for the development of new anthelminthic drugs. AIM OF THE STUDY The purpose of this study was i) to conduct an ethnobotanical survey to gather data on Bangladeshi medicinal plants used in the treatment of helminthiases, ii) to test plants with the highest use values for their in vitro anthelmintic activity, and iii) to carry out in silico screening on phytochemicals present in the most active plant extract to investigate their ability to disrupt β-tubulin function in helminths. METHODS The ethnobotanical survey was conducted across three sub-districts of Bangladesh, namely Mathbaria, Phultala and Khan Jahan Ali. The in vitro screening for anthelmintic activity was performed in a motility test using adult Haemonchus contortus worms. Virtual screening using PyRx was performed on the phytochemicals reported from the most active plant, exploring their interactions with the colchicine binding site of the β-tubulin protein target (PDB ID: 1SA0). RESULTS The survey respondents reported a total of 32 plants for treating helminthiases. Based on their use values, the most popular choices were Ananas comosus (L.) Merr., Azadirachta indica A.Juss., Carica papaya L., Citrus maxima (Burm.) Merr., Curcuma longa L., Momordica charantia L., Nigella sativa L. and Syzygium cumini (L.) Skeels. In vitro anthelmintic testing revealed that A. indica leaves and bark had the highest activity with LC50 values of 16 mg/mL in both cases. Other plant extracts also exhibited good anthelmintic activity with LC50 values ranging from 16 to 52 mg/mL, while the value for albendazole (positive control) was 8.39 mg/mL. The limonoids nimbolide and 28-deoxonimbolide showed a binding affinity of -8.9 kcal/mol, and satisfied all drug-likeness parameters. The control ligand N-deacetyl-N-(2-mercaptoacetyl)colchicine had a binding affinity of -6.9 kcal/mol. CONCLUSION Further in silico and in vitro studies are warranted on the identified limonoids to confirm the potential of these derivatives as novel drug templates for helminthiases. The current study supports the need for an ethnobotanical survey-based approach to discover novel drug templates for helminthiases.
Collapse
Affiliation(s)
- M Khairuzzaman
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Md Mehedi Hasan
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Mohammad Tuhin Ali
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Sheuly Akter
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh; Bangladesh Reference Institute for Chemical Measurements, Dr Kudrat-e-Khuda Road, Dhanmondi, Dhaka, Bangladesh
| | - Papia Nasrin
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Md Khirul Islam
- Department of Life Technologies, Division of Biotechnology, University of Turku, Finland
| | - Akhlak Un Nahar
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Dipto Kumer Sarker
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Omer Abdalla Ahmed Hamdi
- Department of Chemistry, Faculty of Science and Technology, Al-Neelain University, Khartoum, Sudan
| | - Shaikh Jamal Uddin
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Jamil A Shilpi
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh.
| |
Collapse
|