1
|
Zeng R, Chen X, Chen Y, Dong J. FGFR4 inhibition augments paclitaxel-induced cell death in ovarian cancer. Int Immunopharmacol 2025; 155:114626. [PMID: 40245772 DOI: 10.1016/j.intimp.2025.114626] [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: 01/13/2025] [Revised: 03/21/2025] [Accepted: 04/05/2025] [Indexed: 04/19/2025]
Abstract
OBJECTIVES Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy, which has a high mortality rate due to frequent tumor recurrence. The development of drug resistance against the first-line chemotherapeutic agent, such as paclitaxel/Taxol®, represents a critical reason. The mechanisms of paclitaxel resistance remain largely unknown, and druggable drivers which can be targeted to prevent or revert paclitaxel resistance also need to be identified. METHODS Phos-tag-based screens in cells treated with paclitaxel were used to identify key regulators involved in paclitaxel resistance, such as fibroblast growth factor receptor 4 (FGFR4). The functional role of FGFR4 in regulating paclitaxel resistance was further identified using apoptosis assays, which included the identification of apoptotic marker levels and activities. The involvement of FGFR4 downstream signaling pathways involved in paclitaxel resistance were identified through western blotting and quantitative PCR. Their roles in regulating paclitaxel resistance were also validated using apoptosis assays. Immunofluorescent staining was performed to identify the synergy of paclitaxel and FGFR4 inhibition. RESULTS Functional in vitro and in vivo studies demonstrate that FGFR4 depletion suppresses ovarian cancer cell proliferation, migration, and tumor growth. Importantly, FGFR4 silencing or specific inhibition can sensitize ovarian cancer cells to paclitaxel, whereas FGFR4 overexpression confers paclitaxel resistance. Mechanistically, FGFR4 regulates paclitaxel sensitivity in EOC cells through modulating the expression of the anti-apoptotic protein B-cell lymphoma-extra large (Bcl-xL) via MEK-ERK-RSK signaling pathway. The inhibition of Bcl-xL or MEK-ERK-RSK signaling can also enhance paclitaxel-stimulated cytotoxicity. CONCLUSION These findings indicate that targeting FGFR4 can be a promising novel strategy to overcome paclitaxel resistance and improve the outcomes of EOC patients.
Collapse
MESH Headings
- Humans
- Paclitaxel/pharmacology
- Paclitaxel/therapeutic use
- Female
- Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/metabolism
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Animals
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Carcinoma, Ovarian Epithelial/drug therapy
- Mice
- Mice, Nude
- Signal Transduction/drug effects
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
Collapse
Affiliation(s)
- Renya Zeng
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.; Department of Cancer Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China..
| | - Xingcheng Chen
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yuanhong Chen
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA..
| |
Collapse
|
2
|
Li L, Wei P, Kong T, Yuan B, Fu P, Li Y, Wang Y, Zheng J, Wang K. Framework nucleic acid-programmed aptamer-paclitaxel conjugates as targeted therapeutics for triple-negative breast cancer. NANOSCALE HORIZONS 2025; 10:873-884. [PMID: 40078065 DOI: 10.1039/d4nh00652f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Triple-negative breast cancer (TNBC) is highly invasive with a poor prognosis, and chemotherapy remains the clinical treatment of choice. Paclitaxel is a commonly used first-line chemotherapy drug, but its untargeted distribution poses clinical challenges. Inspired by antibody-drug conjugates, we develop a precisely structured framework nucleic acid-programmed aptamer-paclitaxel conjugate (FAPC) with chemically well-defined paclitaxel loading dosing, enabling the regulation of receptor-aptamer affinity to facilitate tumor-targeted chemotherapy. Utilizing framework nucleic acids as a precise addressing scaffold, we organize the AS1411 aptamer with accurate intermolecular spacing and find that an inter-aptamer spacing of 19.04 nm could enhance the affinity of the FAPC for tumor cells. Then, the multifunctional FAPC can disrupt actin reorganization to achieve cytotoxicity in tumor cells. Furthermore, the AS1411-specifically modified FAPC further enhances the structure-dependent selective accumulation of drugs at tumor sites in a human xenograft model of triple-negative breast cancer, subsequently leading to significantly improved antitumor efficacy and reduced toxicity. The FAPC provides a precisely programmable platform for efficient targeted delivery of chemotherapeutic agents to malignancies.
Collapse
Affiliation(s)
- Lin Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Pengyao Wei
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Tong Kong
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Bo Yuan
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Pan Fu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
- St. George and Sutherland Clinical Campuses School of Clinical Medicine UNSW Sydney Kensington, NSW 2052, Australia
| | - Yuhui Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jianping Zheng
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Kaizhe Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
3
|
Kang JH, Cho YJ, Hwang JY, Park SY, Choi JJ, Paik ES, Kim HS, Lee JW, Shin US. Temperature-Controlled pNIB/PTX Micelles for Improved Paclitaxel Delivery in Ovarian Cancer Treatment. ACS Biomater Sci Eng 2025; 11:2167-2179. [PMID: 40094480 DOI: 10.1021/acsbiomaterials.4c02060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Paclitaxel (PTX) is a widely used anticancer drug for ovarian cancer treatment, but its clinical application is limited by poor water solubility and dose-limiting toxicities. To overcome these challenges, we developed a thermoresponsive, multistep drug delivery system, pNIB/PTX, designed to improve PTX solubility and provide controlled drug release. The pNIB/PTX-3 complex exhibited an initial rapid drug release phase followed by sustained slow release, optimizing both short-term and long-term therapeutic efficacy. At physiological temperatures, the complex demonstrated a precisely controlled drug release mechanism driven by changes in the polymeric micelle structure. In vitro studies showed that pNIB/PTX-3 significantly enhanced therapeutic effects in human ovarian cancer cell lines HeyA8 and SKOV3ip1, compared to PTX alone. In orthotopic ovarian cancer mouse models, a single intraperitoneal injection of pNIB/PTX-3 led to a substantial reduction in tumor size and prolonged survival. This multistep, thermoresponsive delivery system shows strong potential as a promising therapeutic option for dose-dense ovarian cancer treatments, providing improved drug stability, controlled release, and minimized side effects.
Collapse
Affiliation(s)
- Ji-Hye Kang
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea
| | - Young-Jae Cho
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Ji-Young Hwang
- Convergence Research Division, Korea Carbon Industry Promotion Agency (KCARBON), 110-11 Ballyong-ro, Deokjin-gu, Jeonju 54853, Republic of Korea
| | - Sang-Yu Park
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea
- Department of Nano-biomedical Science BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea
| | - Jung-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - E Sun Paik
- Department of Obstetrics and Gynecology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea
| | - Han-Sem Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Ueon Sang Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea
- Department of Nano-biomedical Science BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea
| |
Collapse
|
4
|
Penjweini R, Link KA, Qazi S, Mattu N, Zuchowski A, Vasta A, Sackett DL, Knutson JR. Low dose Taxol causes mitochondrial dysfunction in actively respiring cancer cells. J Biol Chem 2025; 301:108450. [PMID: 40147771 PMCID: PMC12052845 DOI: 10.1016/j.jbc.2025.108450] [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: 09/13/2024] [Revised: 03/03/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Mitochondrial oxygen consumption, dynamics, and morphology play roles in the occurrence, development, and drug resistance of cancer; thus, they are main targets for many anticancer drugs. Increased mitochondrial oxygen consumption and impaired oxygen delivery creates hypoxia, which influences the balance of metabolic cofactors for biogenesis, disease progression, and response to therapeutics. We therefore investigated the effects of Taxol, a well-known anticancer drug, on mitochondrial respiration (principally via a measure of oxidative phosphorylation versus glycolysis), morphology, and dynamics. The concomitant effects of Taxol on mitochondrial ATP and reactive oxygen species production, mitochondrial membrane potential, radical-induced formation of carbonyl groups, mitochondrial release of cytochrome c, as well as cell cycle were investigated. Cells used in this study include the following: A549 (non-small-cell lung epithelial cancer cell line), A549-ρ0 (mitochondrial DNA-depleted derivative of A549), and BEAS-2B (a noncancer cell line derived from normal bronchial epithelium), as well as PC3 (prostate cancer) and HepG2 (hepatocellular carcinoma); these cell lines are known to have disparate metabolic profiles. Using a multitude of fluorescence-based measurements, we show that Taxol, even at a low dose, still adversely affects mitochondria of actively respiring (aerobic) cancer cells. We find an increase in mitochondrial ROS and cytochrome c release, suppression of ATP production and oxidative phosphorylation, fragmentation of the mitochondrial network, and disruption of mitochondria-microtubule linkage. We find these changes in oxidative, but not glycolytic, cancer cells. Noncancer cells, which are oxidative, do not show these changes.
Collapse
Affiliation(s)
- Rozhin Penjweini
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Katie A Link
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Shureed Qazi
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Nikhil Mattu
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Adam Zuchowski
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Alexandra Vasta
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Dan L Sackett
- Cytoskeletal Dynamics Group, Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA.
| | - Jay R Knutson
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA.
| |
Collapse
|
5
|
Komar ZM, Verkaik NS, Dahmani A, Montaudon E, Kanaar R, Houtsmuller AB, Jager A, Marangoni E, van Gent DC. Development and validation of a functional ex vivo paclitaxel and eribulin sensitivity assay for breast cancer, the REMIT assay. NPJ Breast Cancer 2025; 11:17. [PMID: 39962055 PMCID: PMC11832732 DOI: 10.1038/s41523-025-00734-x] [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: 08/07/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
Breast cancer is the most common cancer amongst women worldwide, however clinically validated chemotherapy response biomarkers that can accurately predict treatment response in patients are largely lacking. Therefore, in this study, functional paclitaxel and eribulin ex vivo sensitivity assays were developed. Patient derived xenograft (PDX) models were used to compare the ex vivo predicted treatment outcome with the sensitivity of mice in vivo. We validated the previously developed sensitivity assay for paclitaxel, which is based on the ratio between replicating (EdU) and mitotic (phospho-Histone H3; pH3) cells as a proxy for blocked mitosis. The assay showed 90% correlation between the ex vivo and in vivo response to paclitaxel treatment in the PDX models. We propose the term REMIT (REplication MITosis) assay and show that it is also a suitable test to predict eribulin sensitivity. The reproducibility of the REMIT assay for paclitaxel and eribulin was determined to be 80% and 83%, respectively. These results justify further clinical validation of the REMIT assay in breast cancer patients.
Collapse
Affiliation(s)
- Zofia M Komar
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Nicole S Verkaik
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Oncode Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Ahmed Dahmani
- Translational Research Department, Institut Curie, PSL University, Paris, France
| | - Elodie Montaudon
- Translational Research Department, Institut Curie, PSL University, Paris, France
| | - Roland Kanaar
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Oncode Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Adriaan B Houtsmuller
- Erasmus Optical Imaging Center and Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, PSL University, Paris, France
| | - Dik C van Gent
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
- Oncode Institute, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
6
|
Su KC, Radul E, Maier NK, Tsang MJ, Goul C, Moodie B, Marescal O, Keys HR, Cheeseman IM. Functional genetics reveals modulators of antimicrotubule drug sensitivity. J Cell Biol 2025; 224:e202403065. [PMID: 39570287 PMCID: PMC11590752 DOI: 10.1083/jcb.202403065] [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: 03/12/2024] [Revised: 09/04/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
Microtubules play essential roles in diverse cellular processes and are important pharmacological targets for treating human disease. Here, we sought to identify cellular factors that modulate the sensitivity of cells to antimicrotubule drugs. We conducted a genome-wide CRISPR/Cas9-based functional genetics screen in human cells treated with the microtubule-destabilizing drug nocodazole or the microtubule-stabilizing drug paclitaxel. We further conducted a focused secondary screen to test drug sensitivity for ∼1,400 gene targets across two distinct human cell lines and to additionally test sensitivity to the KIF11 inhibitor, STLC. These screens defined gene targets whose loss enhances or suppresses sensitivity to antimicrotubule drugs. In addition to gene targets whose loss sensitized cells to multiple compounds, we observed cases of differential sensitivity to specific compounds and differing requirements between cell lines. Our downstream molecular analysis further revealed additional roles for established microtubule-associated proteins and identified new players in microtubule function.
Collapse
Affiliation(s)
- Kuan-Chung Su
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Elena Radul
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Nolan K. Maier
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mary-Jane Tsang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Claire Goul
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Océane Marescal
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Heather R. Keys
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Iain M. Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
7
|
Xu XX, Smith ER. The Second Selectivity of Taxanes to Malignant Cells --- Nuclear Envelope Malleability. J Cancer 2025; 16:1051-1053. [PMID: 39895785 PMCID: PMC11786023 DOI: 10.7150/jca.104809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/09/2024] [Indexed: 02/04/2025] Open
Affiliation(s)
- Xiang-Xi Xu
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Elizabeth R. Smith
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
8
|
Cüce‐Aydoğmuş EM, İnhan‐Garip GA. Investigation of the Effects of Blocking Potassium Channels With 4-Aminopyridine on Paclitaxel Activity in Breast Cancer Cell Lines. Cancer Rep (Hoboken) 2024; 7:e70072. [PMID: 39648339 PMCID: PMC11625685 DOI: 10.1002/cnr2.70072] [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: 02/06/2024] [Revised: 10/23/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Paclitaxel (PTX) has been used as a chemotherapeutic agent for several malignancies, including breast cancer, and efforts to increase the efficiency of PTX are continuous. Previous studies have shown that the voltage-gated K+ channels are over-expressed in breast cancer cell lines; therefore, blocking this type of K+ channel reduces cell proliferation and viability. AIMS In this study, FDA-approved 4-aminopyridine (4-AP), a voltage-gated potassium channel blocker, was used in combination with PTX to improve the anticancer activity of PTX in MCF-7 and MDA-MB-231 cell lines. METHODS AND RESULTS Viability was determined with trypan blue, a clonogenic assay was performed, and the cell cycle was determined with a flow cytometer and immunochemistry. To gain an insight into the mechanism, intracellular K+ concentration, intracellular Ca2+ (calcium) concentration, and transmembrane potential measurements were made with corresponding fluorescent dyes. The apoptotic cell number was determined using Annexin /PI method by flow cytometer. Viability decreased with combination therapy and the clonogenic assay proved decreased colony formation. The apoptotic cell number was increased after treatment with the combination in both cell lines. Cell cycle measurements showed G1 arrest for both MCF-7 and MDA-MB-231 cell lines upon 4-AP treatment. PTX caused G1 arrest in MCF-7 cells and S phase arrest in MDA-MB-231 cells. Combination treatment caused S phase arrest in MCF-7 cells and S phase and G2/M phase arrest in MDA-MB-231 cells. Intracellular K+ concentration was increased after all treatments in both cell lines. Ca2+ concentration was increased significantly after combination treatment. Depolarization in the transmembrane potential was observed after all treatments in both cell lines. CONCLUSION Biophysical parameters like the transmembrane potential and ion fluxes have been defined in cancer progression which can provide new aspects for cancer treatments. This study shows that the combination of 4-AP with PTX is a promising alternative the mechanism of which needs further investigation considering the results obtained for Ca2+, K+, and membrane potential.
Collapse
Affiliation(s)
- Esra M. Cüce‐Aydoğmuş
- School of Medicine, Biophysics DepartmentT.C. Maltepe UniversityMaltepeİstanbulTurkey
| | - G. Ayşe İnhan‐Garip
- School of Medicine, Biophysics DepartmentT.C. Marmara UniversityMaltepeİstanbulTurkey
| |
Collapse
|
9
|
Pospíšilová J, Heger T, Kurka O, Kvasnicová M, Chládková A, Nemec I, Rárová L, Cankař P. Atropisomeric 1-phenylbenzimidazoles affecting microtubule organization: influence of axial chirality. Org Biomol Chem 2024; 22:6966-6980. [PMID: 38988246 DOI: 10.1039/d4ob00863d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Benzimidazoles are frequently used in medicinal chemistry. Their anticancer effect is among the most prominent biological activities exhibited by this scaffold. Although numerous benzimidazole derivatives have been synthesized, possible atropisomerism of ortho-substituted 1-phenylbenzimidazoles has been largely overlooked. The aim of this research was to synthesize a small library of novel atropisomeric benzimidazole derivatives and explore their biological activity in various cancer and normal human cell lines. The new unique structural motif provides an interesting 3D architecture with axial chirality, which further contributes to molecular complexity and specificity. Racemates and their separated atropisomers arrested the cell cycle, caused apoptosis, and affected microtubule organization in cancer cells in vitro at different intensities. Moreover, this phenomenon was also verified by the inhibition of endothelial cell migration. These results showed that (+)-atropisomers, especially 5n, exhibit a stronger effect and show promise as agents for cancer therapy.
Collapse
Affiliation(s)
- Jana Pospíšilová
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic.
| | - Tomáš Heger
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Slechtitelu 27, 77900 Olomouc, Czech Republic.
| | - Ondřej Kurka
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Sciences and Faculty of Science, Palacký University, Slechtitelu 27, Olomouc CZ-77900, Czech Republic
| | - Marie Kvasnicová
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Slechtitelu 27, 77900 Olomouc, Czech Republic.
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Science, Palacký University, Slechtitelu 27, 77900 Olomouc, Czech Republic
| | - Anna Chládková
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic.
| | - Ivan Nemec
- Department of Inorganic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic
| | - Lucie Rárová
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Slechtitelu 27, 77900 Olomouc, Czech Republic.
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Science, Palacký University, Slechtitelu 27, 77900 Olomouc, Czech Republic
| | - Petr Cankař
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic.
| |
Collapse
|
10
|
Tucker JB, Carlsen CL, Scribano CM, Pattaswamy SM, Burkard ME, Weaver BA. CENP-E Inhibition Induces Chromosomal Instability and Synergizes with Diverse Microtubule-Targeting Agents in Breast Cancer. Cancer Res 2024; 84:2674-2689. [PMID: 38832939 PMCID: PMC11326998 DOI: 10.1158/0008-5472.can-23-3332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/09/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
Drugs that perturb microtubules are commonly used to treat breast cancers of all subtypes in both early stage and metastatic disease, but they are effective in only approximately 50% of patients. High concentrations of microtubule-targeting agents can elicit mitotic arrest in cell culture models; however, recent evidence from primary and metastatic breast cancers has revealed that these agents only accumulate at intratumoral levels capable of inducing abnormal multipolar mitotic spindles, not mitotic arrest. Although the maintenance of multipolar spindles can generate cytotoxic rates of chromosomal instability (CIN), focusing of aberrant multipolar spindles into normal bipolar spindles can dramatically reduce CIN and confer resistance to microtubule poisons. Here, we showed that inhibition of the mitotic kinesin centromeric-associated protein-E (CENP-E) overcomes resistance caused by focusing multipolar spindles. Clinically relevant microtubule-targeting agents used a mechanistically conserved pathway to induce multipolar spindles without requiring centrosome amplification. Focusing could occur at any point in mitosis, with earlier focusing conferring greater resistance to antimicrotubule agents. CENP-E inhibition increased CIN on focused spindles by generating chromosomes that remained misaligned at spindle poles during anaphase, which substantially increased death in the resulting daughter cells. CENP-E inhibition synergized with diverse, clinically relevant microtubule poisons to potentiate cell death in cell lines and suppress tumor growth in orthotopic tumor models. These results suggest that primary resistance to microtubule-targeting drugs can be overcome by simultaneous inhibition of CENP-E. Significance: The increased incidence of polar chromosomes induced by inhibition of the mitotic kinesin CENP-E exacerbates chromosomal instability, reduces daughter cell viability, and improves sensitivity to microtubule-targeting therapies.
Collapse
Affiliation(s)
- John B. Tucker
- Cancer Biology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Caleb L. Carlsen
- Cellular and Molecular Biology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Christina M. Scribano
- Cellular and Molecular Pharmacology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Srishrika M. Pattaswamy
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark E. Burkard
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Beth A. Weaver
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
11
|
Zhang Y, Wei S, Zhang Q, Zhang Y, Sun C. Paris saponin VII inhibits triple-negative breast cancer by targeting the MEK/ERK/STMN1 signaling axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155746. [PMID: 38763012 DOI: 10.1016/j.phymed.2024.155746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/28/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a category of breast cancer characterized with high molecular heterogeneity. Owing to the lack of effective therapeutic strategies, patients with TNBC have a poor prognosis. Paris saponin VII (PSⅦ), a steroidal saponin extracted from the rhizome of Trichillium tschonoskii Maxim, exhibits excellent anti-cancer activity in a variety of solid tumors. However, the role and potential mechanism of PSⅦ against TNBC remain unexplored. PURPOSE This study aimed to elucidate the therapeutic effects of PSⅦ against TNBC and explore the potential mechanism of action. METHODS We combined the analysis of public single-cell sequencing data with weighted gene co-expression network analysis (WGCNA) to identity differentially expressed genes (DEGs) that distinguished malignant and normal epithelial cells in TNBC. Subsequently, the biological features of DEGs in TNBC were evaluated. Gene set enrichment analysis (GSEA) was used to define potential pathways associated with the DEGs. The pharmacological activity of PSⅦ for TNBC was evidenced via in vitro and in vivo experiments, and molecular docking, molecular dynamics (MD), surface plasmon resonance (SPR) assay and western blotting were employed to confirm the relative mechanisms. RESULTS Single-cell sequencing and WGCNA revealed STMN1 as a pivotal biomarker of TNBC. STMN1 overexpression in TNBC was associated with poor patient prognosis. GSEA revealed a significant accumulation of STMN1 within the MAPK signaling pathway. Furthermore, In vitro experiments showed that PSⅦ showed significantly suppressive actions on the proliferation, migration and invasion abilities for TNBC cells, while inducing apoptosis. Molecular docking, MD analysis and SPR assay indicated a robust interaction between PSⅦ and the MEK protein. Western blotting revealed that PSⅦ may inhibit tumor progression by suppressing the phosphorylation of MEK1/2 and the downstream phosphorylation of ERK1/2 and STMN1. Intraperitoneal injection of PSⅦ (10 mg/kg) notably reduced tumor growth by 71.26 % in a 4T1 xenograft model. CONCLUSION In our study, the systems biology method was used to identify potential therapeutic targets for TNBC. In vitro and in vivo experiments demonstrated PSⅦ suppresses cancer progression by targeting the MEK/ERK/STMN1 signaling axis. For the first time, the inhibition of STMN1 phosphorylation has been indicated as a possible mechanism for the anticancer effects of PSⅦ. These results emphasize the potential value of PSⅦ as a promising anti-cancer drug candidate for further development in the field of TNBC therapeutics.
Collapse
Affiliation(s)
- Yubao Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250022, China
| | - Shijie Wei
- Department of Oncology, The Affiliated Qingdao Hiser hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao 266071, China
| | - Qinxiang Zhang
- Institute of Integrated Medicine, Qingdao University, Qingdao 266071, China
| | - Yue Zhang
- Institute of Integrated Medicine, Qingdao University, Qingdao 266071, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, Shandong Province 261000, China.
| |
Collapse
|
12
|
van der Westhuyzen A, Ashraf N, Conradie D, Loots L, Kaschula CH, Pelly SC, Frolova LV, Landfair T, Shuster CB, Betancourt T, Kornienko A, van Otterlo WAL. Improved Rigidin-Inspired Antiproliferative Agents with Modifications on the 7-Deazahypoxanthine C7/C8 Ring Systems. J Med Chem 2024; 67:9950-9975. [PMID: 38865195 PMCID: PMC11215747 DOI: 10.1021/acs.jmedchem.3c02473] [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: 12/30/2023] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
To improve their aqueous solubility characteristics, water-solubilizing groups were added to some antiproliferative, rigidin-inspired 7-deazahypoxanthine frameworks after molecular modeling seemed to indicate that structural modifications on the C7 and/or C8 phenyl groups would be beneficial. To this end, two sets of 7-deazahypoxanthines were synthesized by way of a multicomponent reaction approach. It was subsequently determined that their antiproliferative activity against HeLa cells was retained for those derivatives with a glycol ether at the 4'-position of the C8 aryl ring system, while also significantly improving their solubility behavior. The best of these compounds were the equipotent 6-[4-(2-ethoxyethoxy)benzoyl]-2-(pent-4-yn-1-yl)-5-phenyl-1,7-dihydro-4H-pyrrolo[2,3-d]pyrimidin-4-one 33 and 6-[4-(2-ethoxyethoxy)benzoyl]-5-(3-fluorophenyl)-2-(pent-4-yn-1-yl)-1,7-dihydro-4H-pyrrolo[2,3-d]pyrimidin-4-one 59. Similarly to the parent 1, the new derivatives were also potent inhibitors of tubulin assembly. In treated HeLa cells, live cell confocal microscopy demonstrated their impact on microtubulin dynamics and spindle morphology, which is the upstream trigger of mitotic delay and cell death.
Collapse
Affiliation(s)
| | - Naghmana Ashraf
- Department
of Biology, New Mexico State University, Las Cruces ,New Mexico 88003, United States
| | - Daleen Conradie
- Department
of Chemistry and Polymer Science, Stellenbosch
University, Stellenbosch 7600, South Africa
- Department
of Physiological Sciences, Stellenbosch
University, Stellenbosch 7600, South Africa
| | - Leigh Loots
- Department
of Chemistry and Polymer Science, Stellenbosch
University, Stellenbosch 7600, South Africa
| | - Catherine H. Kaschula
- Department
of Chemistry and Polymer Science, Stellenbosch
University, Stellenbosch 7600, South Africa
| | - Stephen C. Pelly
- Department
of Chemistry and Polymer Science, Stellenbosch
University, Stellenbosch 7600, South Africa
- Department
of Chemistry, Emory University, 1515 Dickey Drive ,Atlanta ,Georgia 30322, United States
| | - Liliya V. Frolova
- Department
of Chemistry and Biochemistry, Purdue University, 2101 East Coliseum Blvd. ,Fort Wayne ,Indiana 46805, United States
| | - Taylor Landfair
- Department
of Biology, New Mexico State University, Las Cruces ,New Mexico 88003, United States
| | - Charles B. Shuster
- Department
of Biology, New Mexico State University, Las Cruces ,New Mexico 88003, United States
| | - Tania Betancourt
- Department
of Chemistry and Biochemistry, Texas State
University, San Marcos ,Texas 78666, United States
| | - Alexander Kornienko
- Department
of Chemistry and Biochemistry, Texas State
University, San Marcos ,Texas 78666, United States
| | - Willem A. L. van Otterlo
- Department
of Chemistry and Polymer Science, Stellenbosch
University, Stellenbosch 7600, South Africa
| |
Collapse
|
13
|
Li Y, Bai X, Yang D. Development and Application of Cationic Nile Blue Probes in Live-Cell Super-Resolution Imaging and Specific Targeting to Mitochondria. ACS CENTRAL SCIENCE 2024; 10:1221-1230. [PMID: 38947205 PMCID: PMC11212141 DOI: 10.1021/acscentsci.4c00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/28/2024] [Accepted: 05/09/2024] [Indexed: 07/02/2024]
Abstract
Mitochondria are essential organelles involved in various metabolic processes in eukaryotes. The imaging, targeting, and investigation of cell death mechanisms related to mitochondria have garnered significant interest. Small-molecule fluorescent probes have proven to be robust tools for utilizing light to advance the study of mitochondrial biology. In this study, we present the rational design of cationic Nile blue probes carrying a permanent positive charge for these purposes. The cationic Nile blue probes exhibit excellent mitochondrial permeability, unique solvatochromism, and resistance to oxidation. We observed weaker fluorescence in aqueous solutions compared to lipophilic solvents, thereby minimizing background fluorescence in the cytoplasm. Additionally, we achieved photoredox switching of the cationic Nile blue probes under mild conditions. This enabled us to demonstrate their application for the first time in single-molecule localization microscopy of mitochondria, allowing us to observe mitochondrial fission and fusion behaviors. Compared to conventional cyanine fluorophores, this class of dyes demonstrated prolonged resistance to photobleaching, likely due to their antioxidation properties. Furthermore, we extended the application of cationic Nile blue probes to the mitochondria-specific delivery of taxanes, facilitating the study of direct interactions between the drug and organelles. Our approach to triggering cell death without reliance on microtubule binding provides valuable insights into anticancer drug research and drug-resistance mechanisms.
Collapse
Affiliation(s)
- Yunsheng Li
- School
of Life Sciences, Westlake University, Hangzhou 310024, China
- Morningside
Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Hong Kong 999077, China
| | - Xiaoyu Bai
- Morningside
Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Hong Kong 999077, China
| | - Dan Yang
- School
of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake
Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| |
Collapse
|
14
|
Yeh JY, Chao HC, Hong CL, Hung YC, Tzou FY, Hsiao CT, Li JL, Chen WJ, Chou CT, Tsai YS, Liao YC, Lin YC, Lin S, Huang SY, Kennerson M, Lee YC, Chan CC. A missense mutation in human INSC causes peripheral neuropathy. EMBO Mol Med 2024; 16:1091-1114. [PMID: 38589651 PMCID: PMC11099080 DOI: 10.1038/s44321-024-00062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024] Open
Abstract
PAR3/INSC/LGN form an evolutionarily conserved complex required for asymmetric cell division in the developing brain, but its post-developmental function and disease relevance in the peripheral nervous system (PNS) remains unknown. We mapped a new locus for axonal Charcot-Marie-Tooth disease (CMT2) and identified a missense mutation c.209 T > G (p.Met70Arg) in the INSC gene. Modeling the INSCM70R variant in Drosophila, we showed that it caused proprioceptive defects in adult flies, leading to gait defects resembling those in CMT2 patients. Cellularly, PAR3/INSC/LGN dysfunction caused tubulin aggregation and necrotic neurodegeneration, with microtubule-stabilizing agents rescuing both morphological and functional defects of the INSCM70R mutation in the PNS. Our findings underscore the critical role of the PAR3/INSC/LGN machinery in the adult PNS and highlight a potential therapeutic target for INSC-associated CMT2.
Collapse
Affiliation(s)
- Jui-Yu Yeh
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - Hua-Chuan Chao
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Neurology, Department of Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Cheng-Li Hong
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - Yu-Chien Hung
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - Fei-Yang Tzou
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - Cheng-Tsung Hsiao
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jeng-Lin Li
- Ph.D. Program in Translational Medicine, National Taiwan University and Academia Sinica, Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital Jinshan Branch, New Taipei City, Taiwan
| | - Wen-Jie Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Tapiei, Taiwan
| | - Cheng-Ta Chou
- Department of Neurology, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yu-Shuen Tsai
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chu Liao
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu, Taiwan
- Department of Medical Science, National Tsing Hua University, HsinChu, Taiwan
| | - Suewei Lin
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Shu-Yi Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Marina Kennerson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney Local Health District, Concord, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Molecular Medicine Laboratory, Concord Hospital, Concord, NSW, Australia
| | - Yi-Chung Lee
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan.
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.
- Brain Research Center, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan.
| | - Chih-Chiang Chan
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
15
|
Lu K, Ye X, Chen Y, Wang P, Gong M, Xuan B, Tang Z, Li M, Hou J, Peng K, Pei H. Research progress of drug eluting balloon in arterial circulatory system. Front Cardiovasc Med 2024; 11:1287852. [PMID: 38601040 PMCID: PMC11005962 DOI: 10.3389/fcvm.2024.1287852] [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: 09/02/2023] [Accepted: 03/04/2024] [Indexed: 04/12/2024] Open
Abstract
The arterial circulatory system diseases are common in clinical practice, and their treatment options have been of great interest due to their high morbidity and mortality. Drug-eluting balloons, as a new type of endovascular interventional treatment option, can avoid the long-term implantation of metal stents and is a new type of angioplasty without stents, so drug-eluting balloons have better therapeutic effects in some arterial circulatory diseases and have been initially used in clinical practice. In this review, we first describe the development, process, and mechanism of drug-eluting balloons. Then we summarize the current studies on the application of drug-eluting balloons in coronary artery lesions, in-stent restenosis, and peripheral vascular disease. As well as the technical difficulties and complications in the application of drug-eluting balloons and possible management options, in order to provide ideas and help for future in-depth studies and provide new strategies for the treatment of more arterial system diseases.
Collapse
Affiliation(s)
- Keji Lu
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- School of Medical and Life Sciences, Chengdu University of TCM, Chengdu, China
| | - Xianglin Ye
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, China
| | - Yaoxuan Chen
- School of Medical and Life Sciences, Chengdu University of TCM, Chengdu, China
| | - Peng Wang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, China
| | - Meiting Gong
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, China
| | - Bing Xuan
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, China
| | - Zhaobing Tang
- Department of Rehabilitation, The General Hospital of Western Theater Command, Chengdu, China
| | - Meiling Li
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, China
| | - Jun Hou
- Department of Cardiology, Chengdu Third People's Hospital, Chengdu, China
| | - Ke Peng
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, China
| | - Haifeng Pei
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
16
|
Hu J, Zhu BY, Niu ZX. Catalysts of Healing: A Symphony of Synthesis and Clinical Artistry in Small-Molecule Agents for Breast Cancer Alleviation. Molecules 2024; 29:1166. [PMID: 38474678 DOI: 10.3390/molecules29051166] [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: 01/11/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Breast cancer, characterized by its molecular intricacy, has witnessed a surge in targeted therapeutics owing to the rise of small-molecule drugs. These entities, derived from cutting-edge synthetic routes, often encompassing multistage reactions and chiral synthesis, target a spectrum of oncogenic pathways. Their mechanisms of action range from modulating hormone receptor signaling and inhibiting kinase activity, to impeding DNA damage repair mechanisms. Clinical applications of these drugs have resulted in enhanced patient survival rates, reduction in disease recurrence, and improved overall therapeutic indices. Notably, certain molecules have showcased efficacy in drug-resistant breast cancer phenotypes, highlighting their potential in addressing treatment challenges. The evolution and approval of small-molecule drugs have ushered in a new era for breast cancer therapeutics. Their tailored synthetic pathways and defined mechanisms of action have augmented the precision and efficacy of treatment regimens, paving the way for improved patient outcomes in the face of this pervasive malignancy. The present review embarks on a detailed exploration of small-molecule drugs that have secured regulatory approval for breast cancer treatment, emphasizing their clinical applications, synthetic pathways, and distinct mechanisms of action.
Collapse
Affiliation(s)
- Jing Hu
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Bi-Yue Zhu
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
| | - Zhen-Xi Niu
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| |
Collapse
|
17
|
Sati P, Sharma E, Dhyani P, Attri DC, Rana R, Kiyekbayeva L, Büsselberg D, Samuel SM, Sharifi-Rad J. Paclitaxel and its semi-synthetic derivatives: comprehensive insights into chemical structure, mechanisms of action, and anticancer properties. Eur J Med Res 2024; 29:90. [PMID: 38291541 PMCID: PMC10826257 DOI: 10.1186/s40001-024-01657-2] [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: 11/09/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
Cancer is a disease that can cause abnormal cell growth and can spread throughout the body. It is among the most significant causes of death worldwide, resulting in approx. 10 million deaths annually. Many synthetic anticancer drugs are available, but they often come with side effects and can interact negatively with other medications. Additionally, many chemotherapy drugs used for cancer treatment can develop resistance and harm normal cells, leading to dose-limiting side effects. As a result, finding effective cancer treatments and developing new drugs remains a significant challenge. However, plants are a potent source of natural products with the potential for cancer treatment. These biologically active compounds may be the basis for enhanced or less toxic derivatives. Herbal medicines/phytomedicines, or plant-based drugs, are becoming more popular in treating complicated diseases like cancer due to their effectiveness and are a particularly attractive option due to their affordability, availability, and lack of serious side effects. They have broad applicability and therapeutic efficacy, which has spurred scientific research into their potential as anticancer agents. This review focuses on Paclitaxel (PTX), a plant-based drug derived from Taxus sp., and its ability to treat specific tumors. PTX and its derivatives are effective against various cancer cell lines. Researchers can use this detailed information to develop effective and affordable treatments for cancer.
Collapse
Affiliation(s)
- Priyanka Sati
- Department of Biotechnology, Kumaun University, Bhimtal, Uttarakhand, India
| | - Eshita Sharma
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Praveen Dhyani
- Institute for Integrated Natural Sciences, University of Koblenz, Koblenz, Germany
| | - Dharam Chand Attri
- Department of Botany, Central University of Jammu, Rahya-Suchani (Bagla), Jammu and Kashmir, India
| | - Rohit Rana
- Department of Biology, Brandeis University, Waltham, MA, USA
| | - Lashyn Kiyekbayeva
- Department of Pharmaceutical Technology, Pharmaceutical School, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar.
| | | |
Collapse
|
18
|
El-Sayed ASA, Shindia A, Ammar H, Seadawy MG, Khashana SA. Bioprocessing of Epothilone B from Aspergillus fumigatus under solid state fermentation: Antiproliferative activity, tubulin polymerization and cell cycle analysis. BMC Microbiol 2024; 24:43. [PMID: 38291363 PMCID: PMC10829302 DOI: 10.1186/s12866-024-03184-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
Epothilone derivatives have been recognized as one of the most powerful anticancer drugs towards solid tumors, for their unique affinity to bind with β-tubulin microtubule arrays, stabilizing their disassembly, causing cell death. Sornagium cellulosum is the main source for Epothilone, however, the fermentation bioprocessing of this myxobacteria is the main challenge for commercial production of Epothilone. The metabolic biosynthetic potency of epothilone by Aspergillus fumigatus, an endophyte of Catharanthus roseus, raises the hope for commercial epothilone production, for their fast growth rate and feasibility of manipulating their secondary metabolites. Thus, nutritional optimization of A. fumigatus for maximizing their epothilone productivity under solid state fermentation process is the objective. The highest yield of epothilone was obtained by growing A. fumigatus on orange peels under solid state fermentation (2.2 μg/g), bioprocessed by the Plackett-Burman design. The chemical structure of the extracted epothilone was resolved from the HPLC and LC-MS/MS analysis, with molecular mass 507.2 m/z and identical molecular fragmentation pattern of epothilone B of S. cellulosum. The purified A. fumigatus epothilone had a significant activity towards HepG2 (IC50 0.98 μg/ml), Pancl (IC50 1.5 μg/ml), MCF7 (IC50 3.7 μg/ml) and WI38 (IC50 4.6 μg/ml), as well as a strong anti-tubulin polymerization activity (IC50 0.52 μg/ml) compared to Paclitaxel (2.0 μg/ml). The effect of A. fumigatus epothilone on the immigration ability of HepG2 cells was assessed, as revealed from the wound closure of the monolayer cells that was estimated by ~ 63.7 and 72.5%, in response to the sample and doxorubicin, respectively, compared to negative control. From the Annexin V-PI flow cytometry results, a significant shift of the normal cells to the apoptosis was observed in response to A. fumigatus epothilone by ~ 20 folds compared to control cells, with the highest growth arrest of the HepG2 cells at the G0-G1 stage.
Collapse
Affiliation(s)
- Ashraf S A El-Sayed
- Enzymology and Fungal Biotechnology lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt.
| | - Ahmed Shindia
- Enzymology and Fungal Biotechnology lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| | - Hala Ammar
- Enzymology and Fungal Biotechnology lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed G Seadawy
- Biological Prevention Department, Egyptian Ministry of Defense, Cairo, Egypt
| | - Samar A Khashana
- Enzymology and Fungal Biotechnology lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
19
|
Elsibaei SM, Amleh A, Ismail MA, El-Sayed WM. Azafuramidines as potential anticancer Agents: Pro-apoptotic profile and cell cycle arrest. Bioorg Med Chem Lett 2024; 97:129550. [PMID: 37952598 DOI: 10.1016/j.bmcl.2023.129550] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/14/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
The current study aimed to test the antiproliferative activity of three azafuramidines (X, Y, and Z) against three different human cell lines; liver HepG2, breast MCF-7, and bone U2OS. And to explore the molecular mechanism(s) of the antiproliferative activity of these derivatives. The three new azafuramidines demonstrated a potent cytotoxicity at < 2 μM against the three cell lines investigated. The azafuramidines were highly selective with selectivity index ∼ 47 - 61 folds indicating safety to the normal cells. In the scratch assay, azafuramidines significantly reduced the percentage of wound healing indicating ability to prevent or reduce metastasis. Derivatives X and Z arrested the HepG2 cells at S and G2/M phases detected by the flow cytometry. Derivatives X, Y, and Z elevated the apoptosis of HepG2 cells by ∼ 71 %, 66 %, and 59 %, respectively. Derivatives X and Z were superior to derivative Y. The potent antiproliferative, cell cycle arrest, and pro-apoptotic efficacy of these chlorophenyl derivatives could be attributed to their ability of inducing the overexpression of p53, p21, and p27. These derivatives had the potential to act as anticancer agents and merit further investigations.
Collapse
Affiliation(s)
- Sameh M Elsibaei
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Asma Amleh
- Department of Biology, School of Science and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Mohamed A Ismail
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo 11566, Egypt.
| |
Collapse
|
20
|
Steffes V, MacDonald S, Crowe J, Murali M, Ewert KK, Li Y, Safinya CR. Lipids with negative spontaneous curvature decrease the solubility of the cancer drug paclitaxel in liposomes. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:128. [PMID: 38099960 PMCID: PMC10802834 DOI: 10.1140/epje/s10189-023-00388-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023]
Abstract
Paclitaxel (PTX) is a hydrophobic small-molecule cancer drug that loads into the membrane (tail) region of lipid carriers such as liposomes and micelles. The development of improved lipid-based carriers of PTX is an important objective to generate chemotherapeutics with fewer side effects. The lipids 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and glyceryl monooleate (GMO) show propensity for fusion with other lipid membranes, which has led to their use in lipid vectors of nucleic acids. We hypothesized that DOPE and GMO could enhance PTX delivery to cells through a similar membrane fusion mechanism. As an important measure of drug carrier performance, we evaluated PTX solubility in cationic liposomes containing GMO or DOPE. Solubility was determined by time-dependent kinetic phase diagrams generated from direct observations of PTX crystal formation using differential-interference-contrast optical microscopy. Remarkably, PTX was much less soluble in these liposomes than in control cationic liposomes containing univalent cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC), which are not fusogenic. In particular, PTX was not substantially soluble in GMO-based cationic liposomes. The fusogenicity of DOPE and GMO is related to the negative spontaneous curvature of membranes containing these lipids, which drives formation of nonlamellar self-assembled phases (inverted hexagonal or gyroid cubic). To determine whether PTX solubility is governed by lipid membrane structure or by local intermolecular interactions, we used synchrotron small-angle X-ray scattering. To increase the signal/noise ratio, we used DNA to condense the lipid formulations into lipoplex pellets. The results suggest that local intermolecular interactions are of greater importance and that the negative spontaneous curvature-inducing lipids DOPE and GMO are not suitable components of liposomal carriers for PTX delivery.
Collapse
Affiliation(s)
- Victoria Steffes
- Materials Department, University of California, Santa Barbara, CA, 93106, USA
- Chemistry and Biochemistry Department, University of California, Santa Barbara, CA, 93106, USA
| | - Scott MacDonald
- Physics Department, University of California, Santa Barbara, CA, 93106, USA
| | - John Crowe
- Physics Department, University of California, Santa Barbara, CA, 93106, USA
| | - Meena Murali
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA, 93106, USA
| | - Kai K Ewert
- Materials Department, University of California, Santa Barbara, CA, 93106, USA
| | - Youli Li
- Materials Research Laboratory, University of California, Santa Barbara, CA, 93106, USA
| | - Cyrus R Safinya
- Materials Department, University of California, Santa Barbara, CA, 93106, USA.
- Physics Department, University of California, Santa Barbara, CA, 93106, USA.
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
21
|
Yang X, Liang Z, Luo Y, Yuan X, Cai Y, Yu D, Xing X. Single-cell impedance cytometry of anticancer drug-treated tumor cells exhibiting mitotic arrest state to apoptosis using low-cost silver-PDMS microelectrodes. LAB ON A CHIP 2023; 23:4848-4859. [PMID: 37860975 DOI: 10.1039/d3lc00459g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Chemotherapeutic drugs such as paclitaxel and vinblastine interact with microtubules and thus induce complex cell states of mitosis arrest at the G2/M phase followed by apoptosis dependent on drug exposure time and concentration. Microfluidic impedance cytometry (MIC), as a label-free and high-throughput technology for single-cell analysis, has been applied for viability assay of cancer cells post drug exposure at fixed time and dosage, yet verification of this technique for varied tumor cell states after anticancer drug treatment remains a challenge. Here we present a novel MIC device and for the first time perform impedance cytometry on carcinoma cells exhibiting progressive states of G2/M arrest followed by apoptosis related to drug concentration and exposure time, after treatments with paclitaxel and vinblastine, respectively. Our results from impedance cytometry reveal increased amplitude and negative phase shift at low frequency as well as higher opacity for HeLa cells under G2/M mitotic arrest compared to untreated cells. The cells under apoptosis, on the other hand, exhibit opposite changes in these electrical parameters. Therefore, the impedance features differentiate the HeLa cells under progressive states post anticancer drug treatment. We also demonstrate that vinblastine poses a more potent drug effect than paclitaxel especially at low concentrations. Our device is fabricated using a unique sacrificial layer-free soft lithography process as compared to the existing MIC device, which gives rise to readily aligned parallel microelectrodes made of silver-PDMS embedded in PDMS channel sidewalls with one molding step. Our results uncover the potential of the MIC device, with a fairly simple and low-cost fabrication process, for cellular state screening in anticancer drug therapy.
Collapse
Affiliation(s)
- Xinlong Yang
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Ziheng Liang
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Yuan Luo
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xueyuan Yuan
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Yao Cai
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Duli Yu
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Xiaoxing Xing
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| |
Collapse
|
22
|
Steffes V, MacDonald S, Crowe J, Murali M, Ewert KK, Li Y, Safinya CR. Lipids with negative spontaneous curvature decrease the solubility of the cancer drug paclitaxel in liposomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.563006. [PMID: 37905081 PMCID: PMC10614943 DOI: 10.1101/2023.10.18.563006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Paclitaxel (PTX) is a hydrophobic small-molecule cancer drug that loads into the membrane (tail) region of lipid carriers such as liposomes and micelles. The development of improved lipid-based carriers of PTX is an important objective to generate chemotherapeutics with fewer side effects. The lipids 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and glyceryl monooleate (GMO) show propensity for fusion with other lipid membranes, which has led to their use in lipid vectors of nucleic acids. We hypothesized that DOPE and GMO could enhance PTX delivery to cells through a similar membrane fusion mechanism. As an important measure of drug carrier performance, we evaluated PTX solubility in cationic liposomes containing GMO or DOPE. Solubility was determined by time-dependent kinetic phase diagrams generated from direct observations of PTX crystal formation using differential-interference-contrast optical microscopy. Remarkably, PTX was much less soluble in these liposomes than in control cationic liposomes containing univalent cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC), which are not fusogenic. In particular, PTX was not substantially soluble in GMO-based cationic liposomes. The fusogenicity of DOPE and GMO is related to the negative spontaneous curvature of membranes containing these lipids, which drives formation of nonlamellar self-assembled phases (inverted hexagonal or gyroid cubic). We used synchrotron small-angle x-ray scattering to determine whether PTX solubility is governed by lipid membrane structure (condensed with DNA in pellet form) or by local intermolecular interactions. The results suggest that local intermolecular interactions are of greater importance and that the negative spontaneous curvature-inducing lipids DOPE and GMO are not suitable components of lipid carriers for PTX delivery regardless of carrier structure.
Collapse
Affiliation(s)
- Victoria Steffes
- Materials Department, University of California, Santa Barbara, California 93106, USA
- Chemistry and Biochemistry Department, University of California, Santa Barbara, California 93106, USA
| | - Scott MacDonald
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, USA
| | - John Crowe
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, USA
| | - Meena Murali
- Materials Department, University of California, Santa Barbara, California 93106, USA
| | - Kai K Ewert
- Materials Department, University of California, Santa Barbara, California 93106, USA
| | - Youli Li
- Physics Department, University of California, Santa Barbara, California 93106, USA
| | - Cyrus R Safinya
- Materials Department, University of California, Santa Barbara, California 93106, USA
- Physics Department, University of California, Santa Barbara, California 93106, USA
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, USA
| |
Collapse
|
23
|
Jeon M, Schmitt DL, Kyoung M, An S. Size-Specific Modulation of a Multienzyme Glucosome Assembly during the Cell Cycle. ACS BIO & MED CHEM AU 2023; 3:461-470. [PMID: 37876499 PMCID: PMC10591302 DOI: 10.1021/acsbiomedchemau.3c00037] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 10/26/2023]
Abstract
Enzymes in glucose metabolism have been subjected to numerous studies, revealing the importance of their biological roles during the cell cycle. However, due to the lack of viable experimental strategies for measuring enzymatic activities particularly in living human cells, it has been challenging to address whether their enzymatic activities and thus anticipated glucose flux are directly associated with cell cycle progression. It has remained largely elusive how human cells regulate glucose metabolism at a subcellular level to meet the metabolic demands during the cell cycle. Meanwhile, we have characterized that rate-determining enzymes in glucose metabolism are spatially organized into three different sizes of multienzyme metabolic assemblies, termed glucosomes, to regulate the glucose flux between energy metabolism and building block biosynthesis. In this work, we first determined using cell synchronization and flow cytometric techniques that enhanced green fluorescent protein-tagged phosphofructokinase is adequate as an intracellular biomarker to evaluate the state of glucose metabolism during the cell cycle. We then applied fluorescence single-cell imaging strategies and discovered that the percentage of Hs578T cells showing small-sized glucosomes is drastically changed during the cell cycle, whereas the percentage of cells with medium-sized glucosomes is significantly elevated only in the G1 phase, but the percentage of cells showing large-sized glucosomes is barely or minimally altered along the cell cycle. Should we consider our previous localization-function studies that showed assembly size-dependent metabolic roles of glucosomes, this work strongly suggests that glucosome sizes are modulated during the cell cycle to regulate glucose flux between glycolysis and building block biosynthesis. Therefore, we propose the size-specific modulation of glucosomes as a behind-the-scenes mechanism that may explain functional association of glucose metabolism with the cell cycle and, thereby, their metabolic significance in human cell biology.
Collapse
Affiliation(s)
- Miji Jeon
- Department
of Chemistry and Biochemistry, University
of Maryland Baltimore County (UMBC); 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| | - Danielle L. Schmitt
- Department
of Chemistry and Biochemistry, University
of Maryland Baltimore County (UMBC); 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| | - Minjoung Kyoung
- Department
of Chemistry and Biochemistry, University
of Maryland Baltimore County (UMBC); 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
- Program
in Oncology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| | - Songon An
- Department
of Chemistry and Biochemistry, University
of Maryland Baltimore County (UMBC); 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
- Program
in Oncology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| |
Collapse
|
24
|
Kumar A, Rana R, Saklani R, Kumar M, Yadav PK, Tiwari A, Chourasia MK. Technology Transfer of a Validated RP-HPLC Method for the Simultaneous Estimation of Andrographolide and Paclitaxel in Application to Pharmaceutical Nanoformulation. J Chromatogr Sci 2023:bmad070. [PMID: 37674403 DOI: 10.1093/chromsci/bmad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 06/09/2023] [Accepted: 08/06/2023] [Indexed: 09/08/2023]
Abstract
Many analytical methods are reported for simultaneous estimation of pharmaceutical dosages form. However, only a few are reproducible at an industrial scale. The proposed research aims to establish a technology transfer (TT) protocol between two laboratories (Lab-X, originator) with binary and (Lab-Y, receiver) with quaternary high-performance liquid chromatography (HPLC) system. Thus, utilizing reverse-phase HPLC (RP-HPLC), a robust, sensitive and repeatable analytical method has been developed, validated and TT between two laboratories for simultaneous estimation of Andrographolide (AG) and Paclitaxel (PTX). The method has been developed on a Phenomenex Luna C18 column (150 x 4.6, 5) sustained at 40°C and validated under the International Conference on Harmonisation (ICH) Q2 (R1) regulatory guideline and TT USP chapter 1224. The mobile phase consisted of MilliQ (pH = 3) and a combination of acetonitrile and methanol (1:1) in the ratio 50:50 with a flow rate of 0.45 mL/min, linear gradient elution in both labs. The AG and PTX were detected on the PDA detector at 224 and 227 nm wavelength with retention time of 4.5 ± 0.34 and 8.2 ± 0.02 min and limit of detection was found 0.028 ± 0.004 μg/mL, and 0.028 ± 0.0007 μg/mL, whereas limit of quantification as 0.086 ± 0.011 μg/mL and 0.088 ± 0.0014 μg/mL respectively in both labs. Throughout, this approach we have proved that proposed method is repeatable in both labs, and it can be used to quantify AG and PTX in developed pharmaceutical nano-formulations.
Collapse
Affiliation(s)
- Abhiram Kumar
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur road, Lucknow, UP 226031, India
| | - Rafquat Rana
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur road, Lucknow, UP 226031, India
| | - Ravi Saklani
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur road, Lucknow, UP 226031, India
| | - Madhaw Kumar
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur road, Lucknow, UP 226031, India
| | - Pavan Kumar Yadav
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur road, Lucknow, UP 226031, India
| | - Amrendra Tiwari
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur road, Lucknow, UP 226031, India
| | - Manish Kumar Chourasia
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur road, Lucknow, UP 226031, India
| |
Collapse
|
25
|
Xie J, Han D, Xu S, Zhang H, Li Y, Zhang M, Deng Z, Tian J, Ye Q. An Image-Based High-Throughput and High-Content Drug Screening Method Based on Microarray and Expansion Microscopy. ACS NANO 2023; 17:15516-15528. [PMID: 37548636 DOI: 10.1021/acsnano.3c01865] [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: 08/08/2023]
Abstract
A high-efficiency drug screening method is urgently needed due to the expanding number of potential targets and the extremely long time required to assess them. To date, high throughput and high content have not been successfully combined in image-based drug screening, which is the main obstacle to improve the efficiency. Here, we establish a high-throughput and high-content drug screening method by preparing a superhydrophobic microwell array plate (SMAP) and combining it with protein-retention expansion microscopy (proExM). Primarily, we described a flexible method to prepare the SMAP based on photolithography. Cells were cultured in the SMAP and treated with different drugs using a microcolumn-microwell sandwiching technology. After drug treatment, proExM was applied to realize super-resolution imaging. As a demonstration, a 7 × 7 image array of microtubules was successfully collected within 3 h with 68 nm resolution using this method. Qualitative and quantitative analyses of microtubule and mitochondria morphological changes after drug treatment suggested that more details were revealed after applying proExM, demonstrating the successful combination of high throughput and high content.
Collapse
Affiliation(s)
- Junfang Xie
- Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, School of Physics and TEDA Applied Physics, Nankai University, Tianjin 300071, China
| | - Daobo Han
- Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, School of Physics and TEDA Applied Physics, Nankai University, Tianjin 300071, China
| | - Shuai Xu
- Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, School of Physics and TEDA Applied Physics, Nankai University, Tianjin 300071, China
| | - Haitong Zhang
- Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, School of Physics and TEDA Applied Physics, Nankai University, Tianjin 300071, China
| | - Yonghe Li
- Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, School of Physics and TEDA Applied Physics, Nankai University, Tianjin 300071, China
| | - Mingshan Zhang
- Institute of Modern Optics, Nankai University, Tianjin 300350, China
| | - Zhichao Deng
- Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, School of Physics and TEDA Applied Physics, Nankai University, Tianjin 300071, China
| | - Jianguo Tian
- Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, School of Physics and TEDA Applied Physics, Nankai University, Tianjin 300071, China
| | - Qing Ye
- Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, School of Physics and TEDA Applied Physics, Nankai University, Tianjin 300071, China
| |
Collapse
|
26
|
Mu Y, Wang J, Li G. Sphingosine kinase-2 silencing modulates sensitivity of HT-29 human colorectal cancer cells to paclitaxel-induced cell death through altering apoptosis-related proteins. Chem Biol Drug Des 2023; 102:255-261. [PMID: 36929215 DOI: 10.1111/cbdd.14232] [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: 02/04/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023]
Abstract
The sphingosine kinase-2 (SphK2), a main component of sphingolipid signal transduction, is reported as an innovative therapeutic candidate for cancer treatment. This study was conducted to investigate the suppression of SphK2 in increasing HT-29 colorectal cancer cells sensitivity to paclitaxel via inducing apoptosis and targeting c-FLIPS, MCL-1, and survivin expressions. Cells were transfected with siRNA against SphK2, PTX, or SphK2 activator. Proliferation and apoptosis were evaluated by MTT assay, and trypan blue staining and ELISA, respectively. SphK2, c-FLIPS, MCL-1, and survivin expression were determined by qRT-PCR and Western blotting. SphK2 siRNA increased cell death induced PTX. SphK2 agonist abolished silencing impact on cell survival. We found down-expression of C-FLIPS, MCL-1, and survivin by SphK2 siRNA transfection either alone or in paclitaxel treated cells, as well as elevation in cell apoptosis. Our results showed that SphK2 suppression may be an effective important modality to enhance cell sensitivity to paclitaxel via the induction of apoptosis in colorectal cancers.
Collapse
Affiliation(s)
- Yagang Mu
- Department of General Surgery, Xi'an XD Group Hospital, Xi'an, China
| | - Jian Wang
- Department of Traditional Chinese Medicine, Dezhou Second People's Hospital, Dezhou, China
| | - Guang Li
- Department of General Surgery, Linyi People's Hospital, Linyi, China
| |
Collapse
|
27
|
Dougherty LL, Avasthi P. Determinants of cytoplasmic microtubule depolymerization during ciliogenesis in Chlamydomonas reinhardtii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.07.536038. [PMID: 37066348 PMCID: PMC10104144 DOI: 10.1101/2023.04.07.536038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
At the core of cilia are microtubules which are important for establishing length and assisting ciliary assembly and disassembly; however, another role for microtubule regulation on ciliogenesis lies outside of the cilium. The microtubule cytoskeleton is a highly dynamic structure which polymerizes and depolymerizes rapidly to assist in cellular processes. These processes have been studied across various organisms with chemical as well as genetic perturbations. However, these have generated conflicting data in terms of the role of cytoplasmic microtubules (CytoMTs) and free tubulin dynamics during ciliogenesis. Here we look at the relationship between ciliogenesis and cytoplasmic microtubule dynamics in Chlamydomonas reinhardtii using chemical and mechanical perturbations. We find that not only can stabilized CytoMTs allow for normal ciliary assembly, but high calcium concentrations and low pH-induced deciliation cause CytoMTs to depolymerize separately from ciliary shedding. In addition, we find that ciliary shedding through mechanical shearing, cilia regenerate earlier despite intact CytoMTs. Our data suggests that cytoplasmic microtubules are not a sink for a limiting pool of cytoplasmic tubulin in Chlamydomonas, depolymerization that occurs following deciliation is a consequence rather than a requirement for ciliogenesis, and intact CytoMTs in the cytoplasm and the proximal cilium support more efficient ciliary assembly.
Collapse
Affiliation(s)
- Larissa L Dougherty
- Biochemistry and Cell Biology Department, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire
| | - Prachee Avasthi
- Biochemistry and Cell Biology Department, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire
| |
Collapse
|
28
|
Elfarnawany A, Dehghani F. Time- and Concentration-Dependent Adverse Effects of Paclitaxel on Non-Neuronal Cells in Rat Primary Dorsal Root Ganglia. TOXICS 2023; 11:581. [PMID: 37505547 PMCID: PMC10385404 DOI: 10.3390/toxics11070581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/27/2023] [Accepted: 07/01/2023] [Indexed: 07/29/2023]
Abstract
Paclitaxel is a chemotherapeutic agent used to treat a wide range of malignant tumors. Although it has anti-tumoral properties, paclitaxel also shows significant adverse effects on the peripheral nervous system, causing peripheral neuropathy. Paclitaxel has previously been shown to exert direct neurotoxic effects on primary DRG neurons. However, little is known about paclitaxel's effects on non-neuronal DRG cells. They provide mechanical and metabolic support and influence neuronal signaling. In the present study, paclitaxel effects on primary DRG non-neuronal cells were analyzed and their concentration or/and time dependence investigated. DRGs of Wister rats (6-8 weeks old) were isolated, and non-neuronal cell populations were separated by the density gradient centrifugation method. Different concentrations of Paclitaxel (0.01 µM-10 µM) were tested on cell viability by MTT assay, cell death by lactate dehydrogenase (LDH) assay, and propidium iodide (PI) assay, as well as cell proliferation by Bromodeoxyuridine (BrdU) assay at 24 h, 48 h, and 72 h post-treatment. Furthermore, phenotypic effects have been investigated by using immunofluorescence techniques. Paclitaxel exhibited several toxicological effects on non-neuronal cells, including a reduction in cell viability, an increase in cell death, and an inhibition of cell proliferation. These effects were concentration- and time-dependent. Cellular and nuclear changes such as shrinkage, swelling of cell bodies, nuclear condensation, chromatin fragmentation, retraction, and a loss in processes were observed. Paclitaxel showed adverse effects on primary DRG non-neuronal cells, which might have adverse functional consequences on sensory neurons of the DRG, asking for consideration in the management of peripheral neuropathy.
Collapse
Affiliation(s)
- Amira Elfarnawany
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle (Saale), Germany;
- Zoology Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle (Saale), Germany;
| |
Collapse
|
29
|
Kawabata R, Izawa N, Suzuki T, Nagahisa Y, Nishikawa K, Takahashi M, Nakamura M, Ishiguro A, Katsuya H, Hihara J, Manaka D, Negoro Y, Tsuji A, Takahashi T, Kochi M, Azuma M, Kadowaki S, Michimae H, Sunakawa Y, Ichikawa W, Fujii M. A Multicenter, Phase II Trial of Schedule Modification for Nab-Paclitaxel in Combination with Ramucirumab for Patients with Previously Treated Advanced Gastric or Gastroesophageal Junction Cancer: The B-RAX Trial (JACCRO GC-09). Target Oncol 2023; 18:359-368. [PMID: 37060430 DOI: 10.1007/s11523-023-00961-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND This study investigated whether schedule modification of bi-weekly nanoparticle albumin-bound paclitaxel (nab-PTX) plus ramucirumab (RAM) is efficacious against gastric cancer (GC) or gastroesophageal junction cancer (GJC). PATIENTS AND METHODS Patients with unresectable GC or GJC who were previously treated with fluoropyrimidine-containing regimens received nab-PTX (100 mg/m2) on days 1, 8, and 15 and RAM (8 mg/kg) on days 1 and 15 of a 28-day cycle. Based on the incidence of severe adverse events (AEs) during the first cycle, patients were modified to bi-weekly therapy from the second cycle. The primary endpoint was progression-free survival (PFS) in the bi-weekly therapy population. Based on the hypothesis that bi-weekly nab-PTX plus RAM would improve PFS from 4.5 to 7.0 months, 40 patients were required for power of 0.8 with a one-sided α of 0.05. RESULTS Of the 81 patients enrolled, 47 patients (58%) were assigned to bi-weekly therapy. Patient characteristics were Eastern Cooperative Oncology Group performance status of 1 (19%) and diffuse type (45%). Median PFS was 4.7 months (95% confidence interval [CI] 3.7-5.6 months) and overall response rate was 25% (95% CI 11-39%). Severe AEs of grade 3 or worse were mainly neutropenia (83%) and hypertension (23%). EQ-5D scores were maintained during the treatment. In patients who continued standard-schedule therapy, median PFS was 2.7 months (95% CI 1.8-4.0 months). CONCLUSIONS The primary endpoint for PFS was statistically not met, but modification of nab-PTX plus RAM to a bi-weekly schedule might be a feasible treatment option as second-line treatment for advanced GC/GJC patients, especially elderly patients, with severe AEs during the first cycle.
Collapse
Affiliation(s)
| | - Naoki Izawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Takahisa Suzuki
- Department of Surgery, National Hospital Organization Kure Medical Center, Kure, Japan
| | - Yoshio Nagahisa
- Department of Surgery, Kurashiki Central Hospital, Kurashiki, Japan
| | - Kazuhiro Nishikawa
- Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Masazumi Takahashi
- Division of Gastroenterological Surgery, Yokohama Municipal Citizen's Hospital, Yokohama, Japan
| | - Masato Nakamura
- Aizawa Comprehensive Cancer Center, Aizawa Hospital, Matsumoto, Japan
| | - Atsushi Ishiguro
- Department of Medical Oncology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Hiroo Katsuya
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Jun Hihara
- Department of Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Dai Manaka
- Department of Surgery, Gastro-Intestinal Center, Kyoto Katsura Hospital, Kyoto, Japan
| | - Yuji Negoro
- Department of Oncological Medicine, Kochi Health Sciences Center, Kochi, Japan
| | - Akihito Tsuji
- Department of Clinical Oncology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takao Takahashi
- Department of Gastroenterological Surgery/Pediatric Surgery, Gifu University, Graduate School of Medicine, Gifu, Japan
| | - Mitsugu Kochi
- Department of Gastrointestinal Surgery, International University of Health and Welfare Ichikawa Hospital, Ichikawa, Japan
| | - Mizutomo Azuma
- Department of Gastroenterology, Kitasato University Hospital, Sagamihara, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hirofumi Michimae
- Department of Clinical Medicine (Biostatistics), School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Wataru Ichikawa
- Division of Medical Oncology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Masashi Fujii
- Department of Digestive Surgery, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
Rezaei N, Kazem Arki M, Miri-Lavasani Z, Solhi R, Khoramipour M, Rashedi H, Asadzadeh Aghdaei H, Hossein-Khannazer N, Mostafavi E, Vosough M. Co-delivery of Doxorubicin and Paclitaxel via Noisome Nanocarriers Attenuates Cancerous Phenotypes in Gastric Cancer Cells. Eur J Pharm Biopharm 2023:S0939-6411(23)00102-9. [PMID: 37105361 DOI: 10.1016/j.ejpb.2023.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Gastric cancer (GC) is known as a deadly malignancy all over the world, yet none of the current therapeutic regimens have achieved efficacy. this current study has aimed to optimize and reduce treatment doses and overcome multidrug resistance in GC by developing optimum niosomal formulation for the delivery of doxorubicin (DXR), paclitaxel (PTX), and their co-delivery. The particles' size, polydispersity index (PDI), and entrapment efficacy (EE%) were optimized using statistical techniques, i.e., Box-Behnken and Central Composite Design. In contrast to soluble drug formulations, the release rate of medicines from nanoparticles were higher in physiological and acidic pH. Niosomes were more stable at 4°C, compared to 25°C. The MTT assay revealed that the IC50 of drug-loaded niosomes was the lowest among all developed formulations. The apoptosis-related genes (CASPASE-3, CASPASE-8, and CASPASE-9) and tumor suppressor genes (BAX, BCL2) were evaluated in cancer cells before and after treatment. In comparison to control cells and cells treated with soluble forms of DXR and PTX, while the expression of BCL2 decreased, the expression of BAX, CASPASE-3, CASPASE-8, and CASPASE-9 was enhanced in cells treated with drug-loaded niosomes. Drug-loaded niosomes inhibited colony formation capacity and increased apoptosis in human AGS gastric cancer cells. Our results indicate that co-delivery of DXR and PTX-loaded niosomes may be an effective and innovative therapeutic approach to gastric cancer.
Collapse
Affiliation(s)
- Niloofar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mandana Kazem Arki
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohre Miri-Lavasani
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Solhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Khoramipour
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Rashedi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
31
|
Leong SW, Wang J, Okuda KS, Su Q, Zhang Y, Abas F, Chia SL, Yusoff K. Discovery of a novel dual functional phenylpyrazole-styryl hybrid that induces apoptotic and autophagic cell death in bladder cancer cells. Eur J Med Chem 2023; 254:115335. [PMID: 37098306 DOI: 10.1016/j.ejmech.2023.115335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/27/2023]
Abstract
Unpleasant side effects and resistance development remained the Achilles heel of chemotherapy. Since low tumor-selectivity and monotonous effect of chemotherapy are closely related to such bottleneck, targeting tumor-selective multi-functional anticancer agents may be an ideal strategy in the search of new safer drugs. Herein, we report the discovery of compound 21, a nitro-substituted 1,5-diphenyl-3-styryl-1H-pyrazole that possesses dual functional characteristics. The 2D- and 3D-culture-based studies revealed that 21 not only could induce ROS-independent apoptotic and EGFR/AKT/mTOR-mediated autophagic cell deaths in EJ28 cells simultaneously but also has the ability in inducing cell death at both proliferating and quiescent zones of EJ28 spheroids. The molecular modelling analysis showed that 21 possesses EGFR targeting capability as it forms stable interactions in the EGFR active site. Together with its good safety profile in the zebrafish-based model, the present study showed that 21 is promising and may lead to the discovery of tumor-selective multi-functional anti-cancer agents.
Collapse
Affiliation(s)
- Sze Wei Leong
- Department of Chemistry, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - JingJing Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Kazuhide Shaun Okuda
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Australia; Department of Anatomy and Physiology and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Qi Su
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia; Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia
| | - Suet Lin Chia
- Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia; Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia; Malaysia Genome and Vaccine Institute (MGVI), National Institute of Biotechnology Malaysia (NIBM), Jalan Bangi, 43000, Kajang, Selangor, Malaysia.
| | - Khatijah Yusoff
- Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia; Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia; Malaysia Genome and Vaccine Institute (MGVI), National Institute of Biotechnology Malaysia (NIBM), Jalan Bangi, 43000, Kajang, Selangor, Malaysia
| |
Collapse
|
32
|
Li ZY, Zhu YX, Chen JR, Chang X, Xie ZZ. The role of KLF transcription factor in the regulation of cancer progression. Biomed Pharmacother 2023; 162:114661. [PMID: 37068333 DOI: 10.1016/j.biopha.2023.114661] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 04/19/2023] Open
Abstract
Kruppel-like factors (KLFs) are a family of zinc finger transcription factors that have been found to play an essential role in the development of various human tissues, including epithelial, teeth, and nerves. In addition to regulating normal physiological processes, KLFs have been implicated in promoting the onset of several cancers, such as gastric cancer, lung cancer, breast cancer, liver cancer, and colon cancer. To inhibit cancer progression, various existing medicines have been used to modulate the expression of KLFs, and anti-microRNA treatments have also emerged as a potential strategy for many cancers. Investigating the possibility of targeting KLFs in cancer therapy is urgently needed, as the roles of KLFs in cancer have not received enough attention in recent years. This review summarizes the factors that regulate KLF expression and function at both the transcriptional and posttranscriptional levels, which could aid in understanding the mechanisms of KLFs in cancer progression. We hope that this review will contribute to the development of more effective anti-cancer medicines targeting KLFs in the future.
Collapse
Affiliation(s)
- Zi-Yi Li
- College of Basic Medical, Nanchang University, Nanchang, Jiangxi 330006, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yu-Xin Zhu
- College of Basic Medical, Nanchang University, Nanchang, Jiangxi 330006, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Jian-Rui Chen
- College of Basic Medical, Nanchang University, Nanchang, Jiangxi 330006, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Xu Chang
- College of Basic Medical, Nanchang University, Nanchang, Jiangxi 330006, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Zhen-Zhen Xie
- College of Basic Medical, Nanchang University, Nanchang, Jiangxi 330006, PR China; Experimental teaching center of Basic Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
33
|
Jiao YT, Jiang H, Wu WT, Qi YT, Wen MY, Yang XK, Kang YR, Zhang XW, Amatore C, Huang WH. Dual-channel nanoelectrochemical sensor for monitoring intracellular ROS and NADH kinetic variations of their concentrations. Biosens Bioelectron 2023; 222:114928. [PMID: 36450163 DOI: 10.1016/j.bios.2022.114928] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022]
Abstract
Reactive oxygen species (ROS) and nicotinamide adenine dinucleotide (NADH) are important intracellular redox-active molecules involved in various pathological processes including inflammation, neurodegenerative diseases, and cancer. However, the fast dynamic changes and mutual regulatory kinetic relationship between intracellular ROS and NADH in these biological processes are still hard to simultaneously investigate. A dual-channel nanowire electrode (DC-NWE) integrating two conductive nanowires, one functionalized with platinum nanoparticles and the other with conductive polymer, was nanofabricated for the selective and simultaneous real-time monitoring of intracellular ROS and NADH release by mitochondria in single living MCF-7 tumoral cells stimulated by resveratrol. The production of ROS was observed to occur tenths of a second before the release of NADH, a significant new piece of information suggesting a mechanism of action of resveratrol. Beyond the importance of the specific data gathered in this study, this work established the feasibility of simultaneously monitoring multiple species and analyzing their kinetics relationships over sub-second time scales thanks to dual-channel nanowire electrodes. It is believed that this concept and its associated nanoelectrochemical tools might benefit to a deeper understanding of mutual regulatory relationship between intracellular crucial molecular markers during physiological and pathological processes as well as for evaluating medical treatments.
Collapse
Affiliation(s)
- Yu-Ting Jiao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Hong Jiang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Wen-Tao Wu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yu-Ting Qi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Ming-Yong Wen
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiao-Ke Yang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yi-Ran Kang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Xin-Wei Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Christian Amatore
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, PR China; PASTEUR, Départment de Chimie, École Normale Supérieure, PSL Research University, Sorbonne University, Paris, 75005, France.
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
34
|
Yoshizawa K, Matsura A, Shimada M, Ishida-Ishihara S, Sato F, Yamamoto T, Yaguchi K, Kawamoto E, Kuroda T, Matsuo K, Tamaoki N, Sakai R, Shimada Y, Mishra M, Uehara R. Tetraploidy-linked sensitization to CENP-E inhibition in human cells. Mol Oncol 2023. [PMID: 36688680 DOI: 10.1002/1878-0261.13379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/23/2022] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Tetraploidy is a hallmark of cancer cells, and tetraploidy-selective cell growth suppression is a potential strategy for targeted cancer therapy. However, how tetraploid cells differ from normal diploids in their sensitivity to anti-proliferative treatments remains largely unknown. In this study, we found that tetraploid cells are significantly more susceptible to inhibitors of a mitotic kinesin (CENP-E) than are diploids. Treatment with a CENP-E inhibitor preferentially diminished the tetraploid cell population in a diploid-tetraploid co-culture at optimum conditions. Live imaging revealed that a tetraploidy-linked increase in unsolvable chromosome misalignment caused substantially longer mitotic delay in tetraploids than in diploids upon moderate CENP-E inhibition. This time gap of mitotic arrest resulted in cohesion fatigue and subsequent cell death, specifically in tetraploids, leading to tetraploidy-selective cell growth suppression. In contrast, the microtubule-stabilizing compound paclitaxel caused tetraploidy-selective suppression through the aggravation of spindle multipolarization. We also found that treatment with a CENP-E inhibitor had superior generality to paclitaxel in its tetraploidy selectivity across a broader spectrum of cell lines. Our results highlight the unique properties of CENP-E inhibitors in tetraploidy-selective suppression and their potential use in the development of tetraploidy-targeting interventions in cancer.
Collapse
Affiliation(s)
- Koya Yoshizawa
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Akira Matsura
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Masaya Shimada
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Sumire Ishida-Ishihara
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Fuyu Sato
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Takahiro Yamamoto
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Kan Yaguchi
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Eiji Kawamoto
- Graduate School of Medicine, Mie University, Tsu, Japan
| | - Taruho Kuroda
- Graduate School of Medicine, Mie University, Tsu, Japan
| | - Kazuya Matsuo
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Kyoto, Japan
| | - Nobuyuki Tamaoki
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
| | - Ryuichi Sakai
- Graduate School and Faculty of Fisheries Sciences, Hokkaido University, Sapporo, Japan
| | - Yasuhito Shimada
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Mithilesh Mishra
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Ryota Uehara
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
35
|
Mondal SK, Jinka S, Shankar G, Srinivas R, Banerjee R. Modification of α-Tocopherol Succinate with a Tumor-targeting Peptide Conjugate Enhances the Antitumor Efficacy of a Paclitaxel-loaded Lipid Aggregate. Chem Asian J 2023; 18:e202201136. [PMID: 36482874 DOI: 10.1002/asia.202201136] [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: 11/09/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Paclitaxel (PTX) is a widely used chemotherapeutic agent in the clinic. However, its clinical benefit is limited due to its low water solubility, off-target toxicity, and for being a multidrug-resistant (MDR) substrate. To overcome these limitations in this study, a tumor-targeting peptide (CRGDK peptide, a ligand for NRP-1 receptor) conjugate of α-tocopheryl succinate (α-TOS) was synthesized and modified on PTX-loaded lipid aggregate (TL-PTX) to leverage the benefits of α-TOS, which include a) anti-cancer activity, b) increased PTX loading, and c) inhibition of MDR activity. Use of peptide conjugate of α-TOS (α-TOS-CRGDK) in lipid aggregate increased PTX entrapment efficiency by 20%, helped in NRP-1 specific cellular uptake and significantly enhanced apoptotic and cell killing activity (p <0.01) of PTX compared to control formulation (CL-PTX) by inhibiting MDR-activity in melanoma resulting in ∼70% increment in overall survival of melanoma tumor-bearing mice. In conclusion, CRGDK- α-TOS conjugate in association with PTX-loaded liposome provided a unique NRP-1 targeted, drug-resistant reversing anticancer regimen for treating aggressive melanoma.
Collapse
Affiliation(s)
- Sujan Kumar Mondal
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad, India
- Department of Radiology, Michigan State University, East Lansing, Michigan (USA
| | - Sudhakar Jinka
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad, India
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gajji Shankar
- Mass Spectrometry Division, CSIR - Indian Institute of Chemical Technology (CSIRIICT), Uppal Road, Tarnaka, Hyderabad, 500 007, Telangana State, India
| | - Ragampeta Srinivas
- Mass Spectrometry Division, CSIR - Indian Institute of Chemical Technology (CSIRIICT), Uppal Road, Tarnaka, Hyderabad, 500 007, Telangana State, India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad, India
| |
Collapse
|
36
|
Meher RK, Pragyandipta P, Reddy PK, Pedaparti R, Kantevari S, Naik PK. Development of 1,3-diynyl derivatives of noscapine as potent tubulin binding anticancer agents for the management of breast cancer. J Biomol Struct Dyn 2022; 40:13136-13153. [PMID: 34583618 DOI: 10.1080/07391102.2021.1982008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We developed 1,3-diynyl derivatives of noscapine (an opium alkaloid) through in silico combinatorial approach and screened out a panel of promising derivatives that bind tubulin and display anticancer activity. The selected derivatives such as 9-4-tBu-Ph-Diyne (20p), 9-3,4-Di-Cl-Diyne (20k) and 9-3,4-Di-F-Diyne (22s) noscapinoids revealed improved predicted binding energy of -6.676 kcal/mol for 20p, -7.294 kcal/mol for 20k and -7.750 kcal/mol for 20s respectively in comparison to noscapine (-5.246 kcal/mol). These 1,3-diynyl derivatives (20p, 29k and 20s) were strategically synthesized in high yields by regioselective modification of noscapine scaffold and HPLC purified (purity is >96%). The decrease in intrinsic fluorescence of purified tubulin to 8.39%, 17.39% and 25.47% by 20p, 20k and 20s respectively, compared to control suggests their binding capability to tubulin. Their cytotoxicity activity was validated based on cellular studies using two human breast adenocarcinoma (MCF-7 and MDA-MB-231), a panel of primary breast tumor cells and one normal human embryonic kidney cell (293 T). The 1,3-diynyl noscapinoids, 20p, 20k and 20s inhibited cellular proliferation in all the cancer cells that ranged between 6.2 and 38.9 µM, without affecting the normal healthy cells (cytotoxicity is <5% at 100 µM). Further, these novel derivatives arrest cell cycle in the G2/M-phase, followed by induction of apoptosis to cancer cells. Thus, we conclude that 1,3-diynyl-noscapinoids have great potential to be a novel therapeutic agent for breast cancers.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rajesh Kumar Meher
- Centre of Excellence in Natural C and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Sambalpur, Odisha, India
| | - Pratyush Pragyandipta
- Centre of Excellence in Natural C and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Sambalpur, Odisha, India
| | - Praveen Kumar Reddy
- Fluoro and Agrochemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Ravikumar Pedaparti
- Fluoro and Agrochemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Srinivas Kantevari
- Fluoro and Agrochemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Pradeep K Naik
- Centre of Excellence in Natural C and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Sambalpur, Odisha, India
| |
Collapse
|
37
|
Sheung JY, Garamella J, Kahl SK, Lee BY, McGorty RJ, Robertson-Anderson RM. Motor-driven advection competes with crowding to drive spatiotemporally heterogeneous transport in cytoskeleton composites. FRONTIERS IN PHYSICS 2022; 10:1055441. [PMID: 37547053 PMCID: PMC10403238 DOI: 10.3389/fphy.2022.1055441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The cytoskeleton-a composite network of biopolymers, molecular motors, and associated binding proteins-is a paradigmatic example of active matter. Particle transport through the cytoskeleton can range from anomalous and heterogeneous subdiffusion to superdiffusion and advection. Yet, recapitulating and understanding these properties-ubiquitous to the cytoskeleton and other out-of-equilibrium soft matter systems-remains challenging. Here, we combine light sheet microscopy with differential dynamic microscopy and single-particle tracking to elucidate anomalous and advective transport in actomyosin-microtubule composites. We show that particles exhibit multi-mode transport that transitions from pronounced subdiffusion to superdiffusion at tunable crossover timescales. Surprisingly, while higher actomyosin content increases the range of timescales over which transport is superdiffusive, it also markedly increases the degree of subdiffusion at short timescales and generally slows transport. Corresponding displacement distributions display unique combinations of non-Gaussianity, asymmetry, and non-zero modes, indicative of directed advection coupled with caged diffusion and hopping. At larger spatiotemporal scales, particles in active composites exhibit superdiffusive dynamics with scaling exponents that are robust to changing actomyosin fractions, in contrast to normal, yet faster, diffusion in networks without actomyosin. Our specific results shed important new light on the interplay between non-equilibrium processes, crowding and heterogeneity in active cytoskeletal systems. More generally, our approach is broadly applicable to active matter systems to elucidate transport and dynamics across scales.
Collapse
Affiliation(s)
- Janet Y. Sheung
- W. M. Keck Science Department, Scripps College, Claremont, CA, United States
- W. M. Keck Science Department, Pitzer College, Claremont, CA, United States
| | - Jonathan Garamella
- Physics and Biophysics Department, University of San Diego, San Diego, CA, United States
| | - Stella K. Kahl
- W. M. Keck Science Department, Scripps College, Claremont, CA, United States
| | - Brian Y. Lee
- W. M. Keck Science Department, Pitzer College, Claremont, CA, United States
| | - Ryan J. McGorty
- Physics and Biophysics Department, University of San Diego, San Diego, CA, United States
| | | |
Collapse
|
38
|
An Overview on Taxol Production Technology and Its Applications as Anticancer Agent. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-022-0063-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
39
|
Smith ER, Wang JQ, Yang DH, Xu XX. Paclitaxel Resistance Related to Nuclear Envelope Structural SturdinessRunning Title: Lamin A/C Expression and Paclitaxel Resistance. Drug Resist Updat 2022; 65:100881. [DOI: 10.1016/j.drup.2022.100881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022]
|
40
|
Ding L, Liu C, Yin S, Zhou Z, Chen J, Chen X, Chen L, Wang D, Liu B, Liu Y, Wei J, Li J. Engineering a dynamic three-dimensional cell culturing microenvironment using a "sandwich" structure-liked microfluidic device with 3D printing scaffold. Biofabrication 2022; 14. [PMID: 35973411 DOI: 10.1088/1758-5090/ac8a19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/16/2022] [Indexed: 11/12/2022]
Abstract
Most of in vivo tissue cells reside in 3D extracellular matrix (ECM) with fluid flow. To better study cell physiology and pathophysiology, there has been an increasing need in the development of methods for culturing cells in in vivo like microenvironments with a number of strategies currently being investigated including hydrogels, spheroids, tissue scaffolds and very promising microfluidic systems. In this paper, a "sandwich" structure-liked microfluidic device integrated with a 3D printing scaffold is proposed for three-dimensional and dynamic cell culture. The device consists of three layers, i.e. upper layer, scaffold layer and bottom layer. The upper layer is used for introducing cells and fixing scaffold, the scaffold layer mimicking ECM is used for providing 3D attachment areas, and the bottom layer mimicking blood vessels is used for supplying dynamic medium for cells. Thermally assisted electrohydrodynamic jet (TAEJ) printing technology and microfabrication technology are combined to fabricate the device. The flow field in the chamber of device is evaluated by numerical simulation and particle tracking technology to investigate the effects of scaffold on fluid microenvironment. The cell culturing processes are presented by the flow behaviours of inks with different colors. The densities and viabilities of HeLa cells are evaluated and compared after 72 h of culturing in the microfluidic devices and 48-well plate. The dose-dependent cell responses to doxorubicin hydrochloride (DOX) are observed after 24 h treatment at different concentrations. These experimental results, including the evaluation of cell proliferation and in vitro cytotoxicity assessment of DOX in the devices and plate, demonstrate that the presented microfluidic device has good biocompatibility and feasibility, which have great potential in providing native microenvironments for in vitro cell studies, tissue engineering and drug screening for tumor therapy.
Collapse
Affiliation(s)
- Laiqian Ding
- Dalian University of Technology, Dalian, Dalian, Liaoning, 116024, CHINA
| | - Chong Liu
- Key Laboratory for Precision and Non-traditional Machining Technology of Ministry of Education, Dalian University of Technology, Dalian, Dalian, Liaoning, 116024, CHINA
| | - Shuqing Yin
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, No.2 Linggong Road, Ganjingzi District, Dalian, Liaoning, 116024, CHINA
| | - Zhanwei Zhou
- Beijing Spacecrafts Co., Ltd., Beijing, Beijing, 100094, CHINA
| | - Jing Chen
- Beijing Spacecrafts Co., Ltd., Beijing, Beijing, 100094, CHINA
| | - Xueting Chen
- Beijing Spacecrafts Co., Ltd., Beijing, Beijing, 100094, CHINA
| | - Li Chen
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, No. 2, Linggong Road, Ganjingzi District, Dalian, Liaoning, 116024, CHINA
| | - Dazhi Wang
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, Dalian, Liaoning, 116024, CHINA
| | - Bo Liu
- Dalian University of Technology, Dalian, Dalian, Liaoning, 116024, CHINA
| | - Yuanchang Liu
- University College London, London, London, London, WC1E 6BT, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Juan Wei
- Centre for Advanced Laser Manufacturing (CALM), School of Mechanical Engineering, Shandong University of Technology, Zibo, Zibo, Shandong, 255049, CHINA
| | - Jingmin Li
- Lab of Biomedical Optics College of Physics and Optoelectronic Engineerin, Dalian University of Technology, Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China, Dalian, 116024, CHINA
| |
Collapse
|
41
|
Andres AE, Mariano A, Rane D, Peterson BR. Quantification of Engagement of Microtubules by Small Molecules in Living Cells by Flow Cytometry. ACS BIO & MED CHEM AU 2022; 2:529-537. [PMID: 36281300 PMCID: PMC9585582 DOI: 10.1021/acsbiomedchemau.2c00031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
Abstract
![]()
Drugs such as paclitaxel (Taxol) that bind microtubules
are widely
used for the treatment of cancer. Measurements of the affinity and
selectivity of these compounds for their targets are largely based
on studies of purified proteins, and only a few quantitative methods
for the analysis of interactions of small molecules with microtubules
in living cells have been reported. We describe here a novel method
for rapidly quantifying the affinities of compounds that bind polymerized
tubulin in living HeLa cells. This method uses the fluorescent molecular
probe Pacific Blue-GABA-Taxol in conjunction with verapamil to block
cellular efflux. Under physiologically relevant conditions of 37 °C,
this combination allowed quantification of equilibrium saturation
binding of this probe to cellular microtubules (Kd = 1.7 μM) using flow cytometry. Competitive binding
of the microtubule stabilizers paclitaxel (cellular Ki = 22 nM), docetaxel (cellular Ki = 16 nM), cabazitaxel (cellular Ki = 6 nM), and ixabepilone (cellular Ki = 10 nM) revealed intracellular affinities for microtubules that
closely matched previously reported biochemical affinities. By including
a cooperativity factor (α) for curve fitting of allosteric modulators,
this probe also allowed quantification of binding (Kb) of the microtubule destabilizers colchicine (Kb = 80 nM, α = 0.08), vinblastine (Kb = 7 nM, α = 0.18), and maytansine (Kb = 3 nM, α = 0.21). Screening of this
assay against 1008 NCI diversity compounds identified NSC 93427 as
a novel microtubule destabilizer (Kb =
485 nM, α = 0.02), illustrating the potential of this approach
for drug discovery.
Collapse
Affiliation(s)
- Angelo E. Andres
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Andres Mariano
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Digamber Rane
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blake R. Peterson
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
42
|
Preparation of size-tunable sub-200 nm PLGA-based nanoparticles with a wide size range using a microfluidic platform. PLoS One 2022; 17:e0271050. [PMID: 35925917 PMCID: PMC9352036 DOI: 10.1371/journal.pone.0271050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/23/2022] [Indexed: 11/19/2022] Open
Abstract
The realization of poly (lactic-co-glycolic acid) nanoparticles (PLGA NPs) from laboratory to clinical applications remains slow, partly because of the lack of precise control of each condition in the preparation process and the rich selectivity of nanoparticles with diverse characteristics. Employing PLGA NPs to establish a large range of size-controlled drug delivery systems and achieve size-selective drug delivery targeting remains a challenge for therapeutic development for different diseases. In this study, we employed a microfluidic device to control the size of PLGA NPs. PLGA, poly (ethylene glycol)-methyl ether block poly (lactic-co-glycolide) (PEG-PLGA), and blend (PLGA + PEG-PLGA) NPs were engineered with defined sizes. Blend NPs exhibit the widest size range (40–114 nm) by simply changing the flow rate conditions without changing the precursor (polymer molecular weight, concentration, and chain segment composition). A model hydrophobic drug, paclitaxel (PTX), was encapsulated in the NPs, and the PTX-loaded NPs maintained a large range of controllable NP sizes. Furthermore, size-controlled NPs were used to investigate the effect of particle size of sub-200 nm NPs on tumor cell growth. The 52 nm NPs showed higher cell growth inhibition than 109 nm NPs. Our method allows the preparation of biodegradable NPs with a large size range without changing polymer precursors as well as the nondemanding fluid conditions. In addition, our model can be applied to elucidate the role of particle sizes of sub-200 nm particles in various biomedical applications, which may help develop suitable drugs for different diseases.
Collapse
|
43
|
Hsa_circ_0011292 regulates paclitaxel resistance partially through regulating CDCA4 expression by serving as a miR-3619-5p sponge in non-small cell lung cancer. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00269-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
44
|
Chmielewski NN, Limoli CL. Sex Differences in Taxane Toxicities. Cancers (Basel) 2022; 14:3325. [PMID: 35884386 PMCID: PMC9317669 DOI: 10.3390/cancers14143325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 02/01/2023] Open
Abstract
The taxane family of microtubule poisons and chemotherapeutics have been studied for over 50 years and are among the most frequently used antineoplastic agents today. Still, limited research exists characterizing taxane-induced sex-specific mechanisms of action and toxicities in cancer and non-cancerous tissue. Such research is important to advance cancer treatment outcomes as well as to address clinically observed sex-differences in short- and long-term taxane-induced toxicities that have disproportionate effects on female and male cancer patients. To gain more insight into these underlying differences between the sexes, the following review draws from pre-clinical and clinical paclitaxel and taxane oncology literature, examines sex-discrepancies, and highlights uncharacterized sex-dependent mechanisms of action and clinical outcomes. To our knowledge, this is the first literature review to provide a current overview of the basic and clinical sex dimorphisms of taxane-induced effects. Most importantly, we hope to provide a starting point for improving and advancing sex-specific personalized chemotherapy and cancer treatment strategies as well as to present a novel approach to review sex as a biological variable in basic and clinical biology.
Collapse
|
45
|
Xu J, Elshazly AM, Gewirtz DA. The Cytoprotective, Cytotoxic and Nonprotective Functional Forms of Autophagy Induced by Microtubule Poisons in Tumor Cells—Implications for Autophagy Modulation as a Therapeutic Strategy. Biomedicines 2022; 10:biomedicines10071632. [PMID: 35884937 PMCID: PMC9312878 DOI: 10.3390/biomedicines10071632] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 12/12/2022] Open
Abstract
Microtubule poisons, as is the case with other antitumor drugs, routinely promote autophagy in tumor cells. However, the nature and function of the autophagy, in terms of whether it is cytoprotective, cytotoxic or nonprotective, cannot be predicted; this likely depends on both the type of drug studied as well as the tumor cell under investigation. In this article, we explore the literature relating to the spectrum of microtubule poisons and the nature of the autophagy induced. We further speculate as to whether autophagy inhibition could be a practical strategy for improving the response to cancer therapy involving these drugs that have microtubule function as a primary target.
Collapse
Affiliation(s)
- Jingwen Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China;
| | - Ahmed M. Elshazly
- Massey Cancer Center, Department of Pharmacology and Toxicology, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - David A. Gewirtz
- Massey Cancer Center, Department of Pharmacology and Toxicology, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
- Correspondence:
| |
Collapse
|
46
|
Anti-Restenotic Technologies in the SFA: Balloons and Stents. Tech Vasc Interv Radiol 2022; 25:100842. [PMID: 35842257 DOI: 10.1016/j.tvir.2022.100842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
Diclofenac: A Nonsteroidal Anti-Inflammatory Drug Inducing Cancer Cell Death by Inhibiting Microtubule Polymerization and Autophagy Flux. Antioxidants (Basel) 2022; 11:antiox11051009. [PMID: 35624874 PMCID: PMC9138099 DOI: 10.3390/antiox11051009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/15/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
Diclofenac, a nonsteroidal anti-inflammatory drug (NSAID) used to treat inflammatory diseases induces cellular toxicity by increasing the production of reactive oxygen species (ROS) and impairing autophagic flux. In this study, we investigated whether diclofenac induces cancer cell death and the mechanism by which diclofenac causes cell death. We observed that diclofenac induces mitotic arrest with a half-maximal effective concentration of 170 μM and cell death with a half-maximal lethal dose of 200 µM during 18-h incubation in HeLa cells. Cellular microtubule imaging and in vitro tubulin polymerization assays demonstrated that treatment with diclofenac elicits microtubule destabilization. Autophagy relies on microtubule-mediated transport and the fusion of autophagic vesicles. We observed that diclofenac inhibits both phagophore movement, an early step of autophagy, and the fusion of autophagosomes and lysosomes, a late step of autophagy. Diclofenac also induces the fragmentation of mitochondria and the Golgi during cell death. We found that diclofenac induces cell death further in combination with 5-fuorouracil, a DNA replication inhibitor than in single treatment in cancer cells. Pancreatic cancer cells, which have high basal autophagy, are particularly sensitive to cell death by diclofenac. Our study suggests that microtubule destabilization by diclofenac induces cancer cell death via compromised spindle assembly checkpoints and increased ROS through impaired autophagy flux. Diclofenac may be a candidate therapeutic drug in certain type of cancers by inhibiting microtubule-mediated cellular events in combination with clinically utilized nucleoside metabolic inhibitors, including 5-fluorouracil, to block cancer cell proliferation.
Collapse
|
48
|
Mishchenko T, Balalaeva I, Gorokhova A, Vedunova M, Krysko DV. Which cell death modality wins the contest for photodynamic therapy of cancer? Cell Death Dis 2022; 13:455. [PMID: 35562364 PMCID: PMC9106666 DOI: 10.1038/s41419-022-04851-4] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
Abstract
Photodynamic therapy (PDT) was discovered more than 100 years ago. Since then, many protocols and agents for PDT have been proposed for the treatment of several types of cancer. Traditionally, cell death induced by PDT was categorized into three types: apoptosis, cell death associated with autophagy, and necrosis. However, with the discovery of several other regulated cell death modalities in recent years, it has become clear that this is a rather simple understanding of the mechanisms of action of PDT. New observations revealed that cancer cells exposed to PDT can pass through various non-conventional cell death pathways, such as paraptosis, parthanatos, mitotic catastrophe, pyroptosis, necroptosis, and ferroptosis. Nowadays, immunogenic cell death (ICD) has become one of the most promising ways to eradicate tumor cells by activation of the T-cell adaptive immune response and induction of long-term immunological memory. ICD can be triggered by many anti-cancer treatment methods, including PDT. In this review, we critically discuss recent findings on the non-conventional cell death mechanisms triggered by PDT. Next, we emphasize the role and contribution of ICD in these PDT-induced non-conventional cell death modalities. Finally, we discuss the obstacles and propose several areas of research that will help to overcome these challenges and lead to the development of highly effective anti-cancer therapy based on PDT.
Collapse
Affiliation(s)
- Tatiana Mishchenko
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Irina Balalaeva
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Anastasia Gorokhova
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Maria Vedunova
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Dmitri V. Krysko
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation ,grid.5342.00000 0001 2069 7798Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium ,grid.510942.bCancer Research Institute Ghent, Ghent, Belgium ,grid.448878.f0000 0001 2288 8774Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| |
Collapse
|
49
|
Hoff KJ, Aiken JE, Gutierrez MA, Franco SJ, Moore JK. Tubulinopathy mutations in TUBA1A that disrupt neuronal morphogenesis and migration override XMAP215/Stu2 regulation of microtubule dynamics. eLife 2022; 11:76189. [PMID: 35511030 PMCID: PMC9236607 DOI: 10.7554/elife.76189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Heterozygous, missense mutations in α- or β-tubulin genes are associated with a wide range of human brain malformations, known as tubulinopathies. We seek to understand whether a mutation’s impact at the molecular and cellular levels scale with the severity of brain malformation. Here, we focus on two mutations at the valine 409 residue of TUBA1A, V409I, and V409A, identified in patients with pachygyria or lissencephaly, respectively. We find that ectopic expression of TUBA1A-V409I/A mutants disrupt neuronal migration in mice and promote excessive neurite branching and a decrease in the number of neurite retraction events in primary rat neuronal cultures. These neuronal phenotypes are accompanied by increased microtubule acetylation and polymerization rates. To determine the molecular mechanisms, we modeled the V409I/A mutants in budding yeast and found that they promote intrinsically faster microtubule polymerization rates in cells and in reconstitution experiments with purified tubulin. In addition, V409I/A mutants decrease the recruitment of XMAP215/Stu2 to plus ends in budding yeast and ablate tubulin binding to TOG (tumor overexpressed gene) domains. In each assay tested, the TUBA1A-V409I mutant exhibits an intermediate phenotype between wild type and the more severe TUBA1A-V409A, reflecting the severity observed in brain malformations. Together, our data support a model in which the V409I/A mutations disrupt microtubule regulation typically conferred by XMAP215 proteins during neuronal morphogenesis and migration, and this impact on tubulin activity at the molecular level scales with the impact at the cellular and tissue levels. Proteins are molecules made up of long chains of building blocks called amino acids. When a mutation changes one of these amino acids, it can lead to the protein malfunctioning, which can have many effects at the cell and tissue level. Given that human proteins are made up of 20 different amino acids, each building block in a protein could mutate to any of the other 19 amino acids, and each mutations could have different effects. Tubulins are proteins that form microtubules, thin tubes that help give cells their shape and allow them to migrate. These proteins are added or removed to microtubules depending on the cell’s needs, meaning that microtubules can grow or shrink depending on the situation. Mutations in the tubulin proteins have been linked to malformations of varying severities involving the formation of ridges and folds on the surface of the brain, including lissencephaly, pachygyria or polymicrogyria. Hoff et al. wanted to establish links between tubulin mutations and the effects observed at both cell and tissue level in the brain. They focused on two mutations in the tubulin protein TUBA1A that affect the amino acid in position 409 in the protein, which is normally a valine. One of the mutations turns this valine into an amino acid called isoleucine. This mutation is associated with pachygyria, which leads to the brain developing few ridges that are broad and flat. The second mutation turns the valine into an alanine, and is linked to lissencephaly, a more severe condition in which the brain develops no ridges, appearing smooth. Hoff et al. found that both mutations interfere with the development of the brain by stopping neurons from migrating properly, which prevents them from forming the folds in the brain correctly. At the cellular level, the mutations lead to tubulins becoming harder to remove from microtubules, making microtubules more stable than usual. This results in longer microtubules that are harder for the cell to shorten or destroy as needed. Additionally, Hoff et al. showed that the mutant versions of TUBA1A have weaker interactions with a protein called XMAP215, which controls the addition of tubulin to microtubules. This causes the microtubules to grow uncontrollably. Hoff et al. also established that the magnitude of the effects of each mutation on microtubule growth scale with the severity of the disorder they cause. Specifically, cells in which TUBA1A is not mutated have microtubules that grow at a normal rate, and lead to typical brain development. Meanwhile, cells carrying the mutation that turns a valine into an alanine, which is linked to the more severe condition lissencephaly, have microtubules that grow very fast. Finally, cells in which the valine is mutated to an isoleucine – the mutation associated with the less severe malformation pachygyria – have microtubules that grow at an intermediate rate. These findings provide a link between mutations in tubulin proteins and larger effects on cell movement that lead to brain malformations. Additionally, they also link the severity of the malformation to the severity of the microtubule defect caused by each mutation. Further work could examine whether microtubule stabilization is also seen in other similar diseases, which, in the long term, could reveal ways to detect and treat these illnesses.
Collapse
Affiliation(s)
- Katelyn J Hoff
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Jayne E Aiken
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Mark A Gutierrez
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Santos J Franco
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Jeffrey K Moore
- University of Colorado School of Medicine, Aurora, United States
| |
Collapse
|
50
|
Spindle motility skews division site determination during asymmetric cell division in Physcomitrella. Nat Commun 2022; 13:2488. [PMID: 35513464 PMCID: PMC9072379 DOI: 10.1038/s41467-022-30239-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 04/21/2022] [Indexed: 01/09/2023] Open
Abstract
Asymmetric cell division (ACD) underlies the development of multicellular organisms. In animal ACD, the cell division site is determined by active spindle-positioning mechanisms. In contrast, it is considered that the division site in plants is determined prior to mitosis by the microtubule-actin belt known as the preprophase band (PPB) and that the localization of the mitotic spindle is typically static and does not govern the division plane. However, in some plant species, ACD occurs in the absence of PPB. Here, we isolate a hypomorphic mutant of the conserved microtubule-associated protein TPX2 in the moss Physcomitrium patens (Physcomitrella) and observe spindle motility during PPB-independent cell division. This defect compromises the position of the division site and produces inverted daughter cell sizes in the first ACD of gametophore (leafy shoot) development. The phenotype is rescued by restoring endogenous TPX2 function and, unexpectedly, by depolymerizing actin filaments. Thus, we identify an active spindle-positioning mechanism that, reminiscent of acentrosomal ACD in animals, involves microtubules and actin filaments, and sets the division site in plants.
Collapse
|