1
|
Zhu N, Wei J, Wang LM, Huang H, Xiao H. Overexpression of PTPN21 promotes proliferation of EGF-stimulated acute lymphoblastic leukemia cells via the MAPK signaling pathways. Hematology 2024; 29:2356292. [PMID: 38785187 DOI: 10.1080/16078454.2024.2356292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
OBJECTIVES This study aims to investigate the role of excessive Protein Tyrosine Phosphatase Non-Receptor Type 21 (PTPN21) in the proliferation of Acute Lymphoblastic Leukemia (ALL) cells with EGF stimulation. METHODS PTPN21 was overexpressed in ALL cell lines by lentiviral transfection. Apoptosis was assayed by Annexin V/7-AAD staining. The proliferation and cell cycle of EGF-treated ALL cells were assessed by MTT and Ki-67/7-AAD staining respectively. The phosphorylation of Src tyrosine kinase and mediators of distinct MAPK pathways were assessed by Western blot. RESULTS Overexpression of PTPN21 had minimal effect on the apoptosis of ALL cells, but significantly promoted the proliferation and cell cycle progression of ALL cells stimulated with EGF. The activity of Src tyrosine kinase and the MAPK pathways was elevated. Inhibition of MAPK pathways by specific inhibitors mitigated this pro-proliferative effect of excessive PTPN21 on EGF-stimulated ALL cells. CONCLUSION PTPN21 may facilitate ALL progression by promoting cell proliferation via the Src/MAPK signaling pathways.
Collapse
Affiliation(s)
- Ni Zhu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, People's Republic of China
| | - Jieping Wei
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Li-Mengmeng Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
| | - Haowen Xiao
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
2
|
Husseiny EM, Abulkhair HS, El-Sebaey SA, Sayed MM, Anwer KE. In vivo evaluation of novel synthetic pyrazolones as CDK9 inhibitors with enhanced pharmacokinetic properties. Future Med Chem 2024:1-19. [PMID: 39530543 DOI: 10.1080/17568919.2024.2419363] [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: 06/22/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
Aim: The structural optimization of our recently reported CDK9 inhibitor to furnish novel aminopyrazolones and methylpyrazolones with improved pharmacokinetics.Materials & methods: The synthesis of the targeted compounds was accomplished via conventional, grinding and microwave-assisted processes. The cytotoxicity of them was assayed against three carcinomas.Results: Analogs 2, 4 and 6 showed significant cytotoxicity and selectivity toward all tested cells. They also displayed potent CDK9 inhibition. Compound 6 arrested MCF-7 cycle at G2/M phase by stimulating the apoptotic pathway. The in vivo biodistribution of radiolabeled compound 6 displayed a potent targeting capability of 131I in solid tumors.Conclusion: Entity 6 is a potent CDK9 inhibitor where 131I-compound 6 can be used as a significant radiopharmaceutical imaging tool for tumors.
Collapse
Affiliation(s)
- Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, 11754, Cairo, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, 11884, Cairo, Egypt
| | - Samiha A El-Sebaey
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, 11754, Cairo, Egypt
| | - Manal M Sayed
- Labeled Compound Department, Hot Labs Center, Egyptian Atomic Energy Authority (EAEA), P. O. Box 13759, Cairo, Egypt
| | - Kurls E Anwer
- Heterocyclic Synthesis Laboratory, Department of Chemistry, Faculty of Science, Ain Shams University 11566, Abbassia, Cairo, Egypt
| |
Collapse
|
3
|
Mondal A, Das B, Karmakar S, Pani S, Khan S, Gupta P, Das Sarma J. Modulatory Role of Pantropic Cell Signaling Pathways in the Antimigratory and Antiproliferative Action of Triazole Chelated Iridium(III) Complexes in Cervical Cancer Cells. J Med Chem 2024. [PMID: 39527836 DOI: 10.1021/acs.jmedchem.4c01997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
In the current study, the antimigratory and antiproliferative effect of three substituted triazole-chelated iridium(III) complexes Ir-TRN, Ir-TRH, and Ir-TRF were studied with special emphasis on modulation of P53 activity, a cell cycle regulator. ERK2/MAPK, another crucial cell signaling pathway protein, was also shown to play a crucial role in cell migration and proliferation. The complexes increase the ROS generation within the cell, further supporting apoptotic induction by exerting cellular oxidative stress. These metal complexes also affect ER stress by altering ERp29, an ER-resident chaperone, further inducing the process of apoptosis. The iridium(III) complexes restrict cervical cancer cell migration and proliferation by exerting pronounced effects as P53 activators and downregulation of ERK2/MAPK activity in cervical cancer cells. The underpinning mechanism of P53 and ERK2/MAPK activity in cervical cancer cells in the presence of iridium(III) complexes was studied in detail in this study, which paves the way for developing promising avenues for cancer therapeutics.
Collapse
Affiliation(s)
- Anushka Mondal
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Bishnu Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Souvik Karmakar
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Soumili Pani
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Shrabani Khan
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Parna Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| |
Collapse
|
4
|
Liu D, Wang H, Chen H, Tian X, Jiao Y, Wang C, Li Y, Li Z, Hou S, Ni Y, Liu B, Lan Y, Zhou J. Ribosome biogenesis is essential for hemogenic endothelial cells to generate hematopoietic stem cells. Development 2024; 151:dev202875. [PMID: 39324287 PMCID: PMC11529273 DOI: 10.1242/dev.202875] [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: 03/14/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Undergoing endothelial-to-hematopoietic transition, a small fraction of embryonic aortic endothelial cells specializes into hemogenic endothelial cells (HECs) and eventually gives rise to hematopoietic stem cells (HSCs). Previously, we found that the activity of ribosome biogenesis (RiBi) is highly enriched in the HSC-primed HECs compared with adjacent arterial endothelial cells; however, whether RiBi is required in HECs for the generation of HSCs remains to be determined. Here, we have found that robust RiBi is markedly augmented during the endothelial-to-hematopoietic transition in mouse. Pharmacological inhibition of RiBi completely impeded the generation of HSCs in explant cultures. Moreover, disrupting RiBi selectively interrupted the HSC generation potential of HECs rather than T1 pre-HSCs, which was in line with its influence on cell cycle activity. Further investigation revealed that, upon HEC specification, the master transcription factor Runx1 dramatically bound to the loci of genes involved in RiBi, thereby facilitating this biological process. Taken together, our study provides functional evidence showing the indispensable role of RiBi in generating HSCs from HECs, providing previously unreported insights that may contribute to the improvement of HSC regeneration strategies.
Collapse
Affiliation(s)
- Di Liu
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China
| | - Haizhen Wang
- The Fifth Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, Guangdong 510900, China
| | - Haifeng Chen
- Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Xitong Tian
- Chinese PLA medical school, Chinese PLA General Hospital, Beijing 100853, China
- State Key Laboratory of Experimental Hematology, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Yuqing Jiao
- Chinese PLA medical school, Chinese PLA General Hospital, Beijing 100853, China
| | - Chi Wang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yuhui Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Zongcheng Li
- State Key Laboratory of Experimental Hematology, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Siyuan Hou
- Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
| | - Yanli Ni
- State Key Laboratory of Experimental Hematology, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Bing Liu
- State Key Laboratory of Experimental Hematology, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yu Lan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jie Zhou
- Chinese PLA medical school, Chinese PLA General Hospital, Beijing 100853, China
- State Key Laboratory of Experimental Hematology, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| |
Collapse
|
5
|
Fahlberg MD, Forward S, Assita ER, Mazzola M, Kiem A, Handley M, Yun SH, Kwok SJJ. Overcoming fixation and permeabilization challenges in flow cytometry by optical barcoding and multi-pass acquisition. Cytometry A 2024; 105:838-848. [PMID: 39467031 DOI: 10.1002/cyto.a.24904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
The fixation and permeabilization of cells are essential for labeling intracellular biomarkers in flow cytometry. However, these chemical treatments often alter fragile targets, such as cell surface and fluorescent proteins (FPs), and can destroy chemically-sensitive fluorescent labels. This reduces measurement accuracy and introduces compromises into sample workflows, leading to losses in data quality. Here, we demonstrate a novel multi-pass flow cytometry approach to address this long-standing problem. Our technique utilizes individual cell barcoding with laser particles, enabling sequential analysis of the same cells with single-cell resolution maintained. Chemically-fragile protein markers and their fluorochrome conjugates are measured prior to destructive sample processing and adjoined to subsequent measurements of intracellular markers after fixation and permeabilization. We demonstrate the effectiveness of our technique in accurately measuring intracellular FPs and methanol-sensitive antigens and fluorophores, along with various surface and intracellular markers. This approach significantly enhances assay flexibility, enabling accurate and comprehensive cellular analysis without the constraints of conventional one-time measurement flow cytometry. This innovation paves new avenues in flow cytometry for a wide range of applications in immuno-oncology, stem cell research, and cell biology.
Collapse
Affiliation(s)
| | | | | | - Michael Mazzola
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Anna Kiem
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Maris Handley
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Seok-Hyun Yun
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
6
|
Guha S, Talukdar D, Mandal GK, Mukherjee R, Ghosh S, Naskar R, Saha P, Murmu N, Das G. Crude extract of Ruellia tuberosa L. flower induces intracellular ROS, promotes DNA damage and apoptosis in triple negative breast cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118389. [PMID: 38821138 DOI: 10.1016/j.jep.2024.118389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ruellia tuberosa L. (Acanthaceae) is a weed plant traditionally used in folklore medicine as a diuretic, anti-hypertensive, anti-pyretic, anti-cancerous, anti-diabetic, analgesic, and gastroprotective agent. It has been previously reported that R. tuberosa L. is enriched with various flavonoids, exhibiting significant cytotoxic potential in various cancer models but a detailed study concerning its molecular mechanism is yet to be explored. AIM OF THE STUDY Exploring and validating R. tuberosa L. flower methanolic extract (RTME) as an anti-cancerous agent as per traditional usage with special emphasis on multi-drug resistant human triple-negative breast cancer (TNBC) and investigating the possible signaling networks and regulatory pathways involved in it. MATERIALS AND METHODS In this study, RTME was prepared using methanol, and phytochemical analysis was performed through GC-MS. Then, the extract was tested for its anti-cancer potential through in-vitro cytotoxicity assay, clonogenic assay, wound healing assay, ROS generation assay, cell cycle arrest, apoptotic nuclear morphology study, cellular apoptosis study, mitochondrial membrane potential (MMP) alteration study, protein, and gene expressions alteration study. In addition, toxicological status was evaluated in female Balb/C mice, and to check the receptor-ligand interactions, in-silico molecular docking was also conducted. RESULTS Several phytochemicals were found within RTME through GC-MS, which have been already reported to act as ROS inductive, DNA damaging, cell cycle arresting, and apoptotic agents against cancer cells. Moreover, RTME was found to exhibit significant in-vitro cytotoxicity along with a reduction in colony formation, and inhibition of cell migratory potential. It also induced intracellular ROS, promoted G0/G1 cell cycle arrest, caused mitochondrial membrane potential (MMP) alteration, and promoted cell death. The Western blot and qRT-PCR data revealed that RTME promoted the intrinsic pathway of apoptosis. Furthermore, blood parameters and organ histology on female Balb/C mice disclosed the non-toxic nature of RTME. Finally, an in-silico molecular docking study displayed that the three identified lead phytochemicals in RTME show strong receptor-ligand interactions with the anti-apoptotic Bcl-2 and give a clue to the possible molecular mechanism of the RTME extract. CONCLUSIONS RTME is a potential source of several phytochemicals that have promising therapeutic potential against TNBC cells, and thus could further be utilized for anti-cancer drug development.
Collapse
Affiliation(s)
- Subhabrata Guha
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India; Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Debojit Talukdar
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Gautam Kumar Mandal
- IQ City Medical College Hospital, IQ City Road, Durgapur, 713206, West Bengal, India.
| | - Rimi Mukherjee
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Srestha Ghosh
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Rahul Naskar
- Department of Chemistry, Jadavpur University, Kolkata, 700032, West Bengal, India.
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Nabendu Murmu
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| | - Gaurav Das
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700026, West Bengal, India.
| |
Collapse
|
7
|
Wang Z, Wang T, Chen X, Lv L, Luo Y, Gu W. ALTMAN: A Novel Method for Cell Cycle Analysis. ACS OMEGA 2024; 9:37780-37788. [PMID: 39281911 PMCID: PMC11391549 DOI: 10.1021/acsomega.4c03653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/27/2024] [Accepted: 07/11/2024] [Indexed: 09/18/2024]
Abstract
Accurate analysis of S-phase fraction is crucial for the assessment of cell proliferation levels, tumor malignancy and prognostic effects of treatment. Most of the currently developed methods for S-phase cell analysis rely on flow cytometric analysis of DNA content determination. However, the lack of standardized procedures for sample analysis and interpretation of cell cycle fitting graphs poses a significant limitation in clinical practice for utilizing flow cytometry to measure the cell cycle based on DNA content. Herein, we developed an approach for analyzing S-phase cells based on telomerase activity determination. Briefly, this approach distinguishes S-phase cells in cell populations via direct fluorescence tracking of telomerase activity within individual cells. The dynamic analysis of telomerase activity in different cell cycles was made possible by the ALTMAN strategy developed in our previous studies, which has been successfully employed to distinguish S-phase cells in cultured cells. This method offers a novel avenue for the assessment of cell cycle status and the evaluation of the proliferation status of tumor cells and the prognosis effect of tumor patients via analyzing the differences in telomerase activity during different cell cycle processes.
Collapse
Affiliation(s)
- Zining Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Tian Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Xiaohui Chen
- Department of Clinical Laboratory, Fuling Hospital, Chongqing University, Chongqing 408099, People's Republic of China
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing University, Chongqing 400044, People's Republic of China
| | - Linxi Lv
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing University, Chongqing 400044, People's Republic of China
| | - Wei Gu
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| |
Collapse
|
8
|
Mehta S, Buyanbat A, Kai Y, Orkin SH. Mechanistic and kinetic insights into transcription factor biology via acute protein depletion. Exp Hematol 2024; 137:104250. [PMID: 38862078 DOI: 10.1016/j.exphem.2024.104250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
Genetic downregulation of the BCL11A transcription factor (TF) reverses the switch from fetal to adult hemoglobin and is effective in treating β-hemoglobinopathies. Genetic ablation results in a gradual reduction in protein abundance and does not lend itself to the analysis of the immediate consequences of protein loss or the determination of the direct interactors/targets of the protein of interest. We achieved acute degradation of the largely disordered and 'undruggable' BCL11A protein by fusing it with a conditional degradation (degron) tag, FKBP12F36V, called degradable tags (dTAG). Small molecules then depleted the BCL11A-dTAG through endogenous proteolytic pathways. By integrating acute depletion with nascent transcriptomics and cell cycle separation techniques, we demonstrate the necessity of BCL11A occupancy at the target chromatin for sustained transcriptional repression in erythroid cells. We advocate for expanding the exploration of TF function to include acute depletion, which holds the potential to unveil unprecedented kinetic insights into TF mechanisms of action.
Collapse
Affiliation(s)
- Stuti Mehta
- Dana Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Altantsetseg Buyanbat
- Dana Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Yan Kai
- Dana Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Stuart H Orkin
- Dana Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA; Boston Children's Hospital, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA.
| |
Collapse
|
9
|
Fahlberg MD, Forward S, Assita ER, Mazzola M, Kiem A, Handley M, Yun SH, Kwok SJJ. Overcoming fixation and permeabilization challenges in flow cytometry by optical barcoding and multi-pass acquisition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607771. [PMID: 39185194 PMCID: PMC11343140 DOI: 10.1101/2024.08.13.607771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
The fixation and permeabilization of cells are essential for labeling intracellular biomarkers in flow cytometry. However, these chemical treatments often alter fragile targets, such as cell surface and fluorescent proteins, and can destroy chemically-sensitive fluorescent labels. This reduces measurement accuracy and introduces compromises into sample workflows, leading to losses in data quality. Here, we demonstrate a novel multi-pass flow cytometry approach to address this long-standing problem. Our technique utilizes individual cell barcoding with laser particles, enabling sequential analysis of the same cells with single-cell resolution maintained. Chemically-fragile protein markers and their fluorochrome conjugates are measured prior to destructive sample processing and adjoined to subsequent measurements of intracellular markers after fixation and permeabilization. We demonstrate the effectiveness of our technique in accurately measuring intracellular fluorescent proteins and methanol-sensitive antigens and fluorophores, along with various surface and intracellular markers. This approach significantly enhances assay flexibility, enabling accurate and comprehensive cell analysis without the constraints of conventional one-time measurement flow cytometry. This innovation paves new avenues in flow cytometry for a wide range of applications in immuno-oncology, stem cell research, and cell biology.
Collapse
Affiliation(s)
| | | | | | - Michael Mazzola
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Anna Kiem
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Maris Handley
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - Seok-Hyun Yun
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA
| | | |
Collapse
|
10
|
Bhattacharjee A, Bagchi A, Sarkar S, Bawali S, Bhattacharya A, Biswas A. Repurposing approved protein kinase inhibitors as potent anti-leishmanials targeting Leishmania MAP kinases. Life Sci 2024; 351:122844. [PMID: 38897344 DOI: 10.1016/j.lfs.2024.122844] [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: 02/23/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
AIMS Leishmaniasis, caused by the protozoan parasite poses a significant health burden globally. With a very few specific drugs, increased drug resistance it is important to look for drug repurposing along with the identification of pre-clinical candidates against visceral leishmaniasis. This study aims to identify potential drug candidates against visceral leishmaniasis by targeting leishmanial MAP kinases and screening FDA approved protein kinase inhibitors. MATERIALS AND METHODS MAP kinases were identified from the Leishmania genome. 12 FDA approved protein kinase inhibitors were screened against Leishmania MAP kinases. Binding affinity, ADME and toxicity of identified drug candidates were profiled. The anti-proliferative effects and mechanism of action were assessed in Leishmania, including changes in cell morphology, flagellar length, cell cycle progression, reactive oxygen species (ROS) generation, and intra-macrophage parasitic burden. KEY FINDINGS 23 MAP kinases were identified from the Leishmania genome. Sorafenib and imatinib emerged as repurposable drug candidates and demonstrated excellent anti-proliferative effects in Leishmania. Treatment with these inhibitors resulted in significant changes in cell morphology, flagellar length, and cell cycle arrest. Furthermore, sorafenib and imatinib promoted ROS generation and reduced intra-macrophage parasitic burden, and elicited anti-leishmanial activity in in vivo experimental VL models. SIGNIFICANCE Collectively, these results imply involvement of MAP kinases in infectivity and survival of the parasite and can pave the avenue for repurposing sorafenib and imatinib as anti-leishmanial agents. These findings contribute to the exploration of new treatment options for visceral leishmaniasis, particularly in the context of emerging drug resistance.
Collapse
Affiliation(s)
- Anindita Bhattacharjee
- Cell and Molecular Biology Laboratory, Department of Zoology, University of Kalyani, Kalyani, Nadia 741235, India
| | - Arka Bagchi
- Cell and Molecular Biology Laboratory, Department of Zoology, University of Kalyani, Kalyani, Nadia 741235, India
| | - Solanki Sarkar
- Cell and Molecular Biology Laboratory, Department of Zoology, University of Kalyani, Kalyani, Nadia 741235, India
| | - Sriparna Bawali
- Cell and Molecular Biology Laboratory, Department of Zoology, University of Kalyani, Kalyani, Nadia 741235, India
| | - Arijit Bhattacharya
- AMR Research Laboratory, Department of Biological Sciences, Adamas University, Kolkata 700126, India.
| | - Arunima Biswas
- Cell and Molecular Biology Laboratory, Department of Zoology, University of Kalyani, Kalyani, Nadia 741235, India.
| |
Collapse
|
11
|
Choithramani A, Das R, Bothra G, Patel Vatsa P, Muthukumar V, Bhuvana BKS, Kapoor S, Moola D, Chowdhury MG, Mandoli A, Shard A. Targeted suppression of oral squamous cell carcinoma by pyrimidine-tethered quinoxaline derivatives. RSC Med Chem 2024; 15:2729-2744. [PMID: 39149105 PMCID: PMC11324040 DOI: 10.1039/d4md00042k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/17/2024] [Indexed: 08/17/2024] Open
Abstract
Oral cancer (OC) stands as a prominent cause of global mortality. Despite numerous efforts in recent decades, the efficacy of novel therapies to extend the lifespan of OC patients remains disappointingly low. Consequently, the demand for innovative therapeutic agents has become all the more pressing. In this context, we present our work on the design and synthesis of twenty-five novel quinoxaline-tethered imidazopyri(mi)dine derivatives. This was followed by comprehensive investigations into the impact of these molecules on the OC cell line. The in vitro cytotoxicity studies performed in CAL-27 and normal oral epithelial (NOE) cell lines revealed that some of the synthesized molecules like 12d have potent antiproliferative activity specifically towards OC cells with an IC50 of 0.79 μM and show negligible cytotoxicity over NOE cells. Further, 12d arrested cell growth in the S phase of the cell cycle and induced cell death by early apoptosis. The in silico studies validated that 12d binds to the activator binding site on pyruvate kinase M2 (PKM2) overexpressed in OC while the lactate dehydrogenase (LDH)-coupled enzyme assay established 12d as a potent PKM2 activator with an AC50 of 0.6 nM. Hence, this study provides fruitful evidence for the designed compounds as anticancer agents against OC.
Collapse
Affiliation(s)
- Asmita Choithramani
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Gourav Bothra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Priyanka Patel Vatsa
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Venkatesh Muthukumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Bombothu Kavya Sai Bhuvana
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Saumya Kapoor
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Deepshika Moola
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Moumita Ghosh Chowdhury
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Amit Mandoli
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| |
Collapse
|
12
|
Sheta YS, Sarg MT, Abdulrahman FG, Nossier ES, Husseiny EM. Novel imidazolone derivatives as potential dual inhibitors of checkpoint kinases 1 and 2: Design, synthesis, cytotoxicity evaluation, and mechanistic insights. Bioorg Chem 2024; 149:107471. [PMID: 38823311 DOI: 10.1016/j.bioorg.2024.107471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Applying various drug design strategies including ring variation, substituents variation, and ring fusion, two series of 2-(alkylthio)-5-(arylidene/heteroarylidene)imidazolones and imidazo[1,2-a]thieno[2,3-d]pyrimidines were designed and prepared as dual potential Chk1 and Chk2 inhibitors. The newly synthesized hybrids were screened in NCI 60 cell line panel where the most active derivatives 4b, d-f, and 6a were further estimated for their five dose antiproliferative activity against the most sensitive tumor cells including breast MCF-7 and MDA-MB-468 and non-small cell lung cancer EKVX as well as normal WI-38 cell. Noticeably, increasing the carbon chain attached to thiol moiety at C-2 of imidazolone scaffold elevated the cytotoxic activity. Hence, compounds 4e and 4f, containing S-butyl fragment, exhibited the most antiproliferative activity against the tested cells where 4f showed extremely potent selectivity toward them. As well, compound 6a, containing imidazothienopyrimidine core, exerted significant cytotoxic activity and selectivity toward the examined cells. The mechanistic investigation of the most active cytotoxic analogs was achieved through the evaluation of their inhibitory activity against Chk1 and Chk2. Results revealed that 4f displayed potent dual inhibition of both Chk1 and Chk2 with IC50 equal 0.137 and 0.25 μM, respectively. It also promoted its antiproliferative and Chk suppression activity via EKVX cell cycle arrest at S phase through stimulating the apoptotic approach. The apoptosis induction was also emphasized by elevating the expression of Caspase-3 and Bax, that are accompanied by Bcl-2 diminution. The in silico molecular docking and ADMET profiles of the most active analogs have been carried out to evaluate their potential as significant anticancer drug candidates.
Collapse
Affiliation(s)
- Yasmin S Sheta
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Marwa T Sarg
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Fatma G Abdulrahman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Eman S Nossier
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt; The National Committee of Drugs, Academy of Scientific Research and Technology, Cairo 11516, Egypt
| | - Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt.
| |
Collapse
|
13
|
Silva-Pavez E, Mendoza E, Morgado-Cáceres P, Ahumada-Castro U, Bustos G, Kangme-Encalada M, de Arbina AL, Puebla-Huerta A, Muñoz F, Cereceda L, Varas-Godoy M, Hidalgo Y, Cardenas JC. Mitochondrial division inhibitor (mdivi-1) induces extracellular matrix (ECM)-detachment of viable breast cancer cells by a DRP1-independent mechanism. Sci Rep 2024; 14:14178. [PMID: 38898058 PMCID: PMC11187114 DOI: 10.1038/s41598-024-64228-9] [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: 02/02/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024] Open
Abstract
Increasing evidence supports the hypothesis that cancer progression is under mitochondrial control. Mitochondrial fission plays a pivotal role in the maintenance of cancer cell homeostasis. The inhibition of DRP1, the main regulator of mitochondrial fission, with the mitochondrial division inhibitor (mdivi-1) had been associated with cancer cell sensitivity to chemotherapeutics and decrease proliferation. Here, using breast cancer cells we find that mdivi-1 induces the detachment of the cells, leading to a bulk of floating cells that conserved their viability. Despite a decrease in their proliferative and clonogenic capabilities, these floating cells maintain the capacity to re-adhere upon re-seeding and retain their migratory and invasive potential. Interestingly, the cell detachment induced by mdivi-1 is independent of DRP1 but relies on inhibition of mitochondrial complex I. Furthermore, mdivi-1 induces cell detachment rely on glucose and the pentose phosphate pathway. Our data evidence a novel DRP1-independent effect of mdivi-1 in the attachment of cancer cells. The generation of floating viable cells restricts the use of mdivi-1 as a therapeutic agent and demonstrates that mdivi-1 effect on cancer cells are more complex than anticipated.
Collapse
Affiliation(s)
- Eduardo Silva-Pavez
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Bellavista, Bellavista 7, Recoleta, Santiago, Chile.
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile.
| | - Elizabeth Mendoza
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Pablo Morgado-Cáceres
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Galdo Bustos
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Matías Kangme-Encalada
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | | | - Andrea Puebla-Huerta
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Felipe Muñoz
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Lucas Cereceda
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Biomedical Research and Innovation Center (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Manuel Varas-Godoy
- Cancer Cell Biology Lab., Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago, Chile
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Avenida Del Valle Norte 725, Huechuraba, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Yessia Hidalgo
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Biomedical Research and Innovation Center (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - J Cesar Cardenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile.
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
- Buck Institute for Research on Aging, Novato, USA.
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, USA.
| |
Collapse
|
14
|
Ou WT, Wan QX, Wu YB, Sun X, Li YL, Tang D, Zhang J, Li SS, Wang NY, Liu ZL, Wu JJ. Long Noncoding RNA PSMB8-AS1 Mediates the Tobacco-Carcinogen-Induced Transformation of a Human Bronchial Epithelial Cell Line by Regulating Cell Cycle. Chem Res Toxicol 2024; 37:957-967. [PMID: 38771128 DOI: 10.1021/acs.chemrestox.4c00025] [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: 05/22/2024]
Abstract
Lung cancer is the main cause of cancer deaths around the world. Nitrosamine 4-(methyl nitrosamine)-1-(3-pyridyl)-1-butanone (NNK) is a tobacco-specific carcinogen of lung cancer. Abundant evidence implicates long noncoding RNAs (lncRNAs) in tumorigenesis. Yet, the effects and mechanisms of lncRNAs in NNK-induced carcinogenesis are still unclear. In this study, we discovered that NNK-induced transformed Beas-2B cells (Beas-2B-NNK) showed increased cell migration and proliferation while decreasing rates of apoptosis. RNA sequencing and differentially expressed lncRNAs analyses showed that lncRNA PSMB8-AS1 was obviously upregulated. Interestingly, silencing the lncRNA PSMB8-AS1 in Beas-2B-NNK cells reduced cell proliferation and migration and produced cell cycle arrest in the G2/M phase along with a decrease in CDK1 expression. Conclusively, our results demonstrate that lncRNA PSMB8-AS1 could promote the malignant characteristics of Beas-2B-NNK cells by regulating CDK1 and affecting the cell cycle, suggesting that it may supply a new prospective epigenetic mechanism for lung cancer.
Collapse
Affiliation(s)
- Wan-Ting Ou
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Qiu-Xian Wan
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yi-Bo Wu
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Xuan Sun
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yan-Li Li
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Dan Tang
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Jian Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Sheng-Sheng Li
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Nuo-Yan Wang
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Zhuo-Lin Liu
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Jian-Jun Wu
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, P. R. China
- State Key Laboratory of Respiratory Disease, Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, P. R. China
| |
Collapse
|
15
|
Abedin Y, Fife A, Samuels CA, Wright R, Murphy T, Zhang X, Alpert E, Cheung E, Zhao Q, Einstein MH, Douglas NC. Combined Treatment of Uterine Leiomyosarcoma with Gamma Secretase Inhibitor MK-0752 and Chemotherapeutic Agents Decreases Cellular Invasion and Increases Apoptosis. Cancers (Basel) 2024; 16:2184. [PMID: 38927890 PMCID: PMC11201464 DOI: 10.3390/cancers16122184] [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: 04/16/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Due to limited effective therapeutics for uterine leiomyosarcoma (uLMS), the impact of the gamma secretase inhibitor (GSI) MK-0752 with common chemotherapeutics was explored in uLMS. MTT assays were performed on two human uLMS cell lines, SK-UT-1B and SK-LMS-1, using MK-0752, docetaxel, doxorubicin, and gemcitabine, individually and in combination, to determine cell viability after treatment. Synergistic combinations were used in transwell invasion assays, cell cycle flow cytometry, proliferation assays, and RNA sequencing. In SK-UT-1B, MK-0752 was synergistic with doxorubicin and gemcitabine plus docetaxel. In SK-LMS-1, MK-0752 was synergistic with all individual agents and with the combination of gemcitabine plus docetaxel. MK-0752, gemcitabine, and docetaxel decreased invasion in SK-UT-1B 2.1-fold* and in SK-LMS-1 1.7-fold*. In SK-LMS-1, invasion decreased 1.2-fold* after treatment with MK-0752 and docetaxel and 2.2-fold* after treatment with MK-0752 and doxorubicin. Cell cycle analysis demonstrated increases in the apoptotic sub-G1 population with MK-0752 alone in SK-UT-1B (1.4-fold*) and SK-LMS-1 (2.7-fold**), along with increases with all combinations in both cell lines. The combination treatments had limited effects on proliferation, while MK-0752 alone decreased proliferation in SK-LMS-1 (0.63-fold**). Both MK-0752 alone and in combination altered gene expression and KEGG pathways. In conclusion, the combinations of MK-0752 with either doxorubicin, docetaxel, or gemcitabine plus docetaxel are potential novel therapeutic approaches for uLMS. (* p < 0.05, ** p < 0.01).
Collapse
Affiliation(s)
- Yasmin Abedin
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Alexander Fife
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Cherie-Ann Samuels
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Rasheena Wright
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Trystn Murphy
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Xusheng Zhang
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA;
| | - Emily Alpert
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Emma Cheung
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Qingshi Zhao
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Mark H. Einstein
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Nataki C. Douglas
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA
| |
Collapse
|
16
|
Chiu CH, Yang DJ, Liou YC, Chang WC, Yu TH, Chung MC, Lee YC, Chen IJ, Wang PY, Lin CP, Tsay HJ, Yeh SHH. Assessment of DNA/RNA Deregulation in Cancer Using 99mTc-Labeled Thiopurine. Cancer Biother Radiopharm 2024; 39:358-372. [PMID: 38457659 DOI: 10.1089/cbr.2023.0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024] Open
Affiliation(s)
- Chuang-Hsin Chiu
- Department of Nuclear Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - David J Yang
- Seecure/Taiwan Hopax Chemicals MFG Company Ltd., Kaohsiung, Taiwan
| | - Yi-Chen Liou
- Institute of Neuroscience, National Chaio Tung University, Taipei, Taiwan
| | - Wei-Chung Chang
- Seecure/Taiwan Hopax Chemicals MFG Company Ltd., Kaohsiung, Taiwan
| | - Tsung-Hsun Yu
- Brain Research Center, National Chaio Tung University, Taipei, Taiwan
- Brain Research Center, School of Medicine, National Defense Medical Center Taipei, Taiwan
| | - Min-Ching Chung
- Seecure/Taiwan Hopax Chemicals MFG Company Ltd., Kaohsiung, Taiwan
| | - Yen-Chun Lee
- Seecure/Taiwan Hopax Chemicals MFG Company Ltd., Kaohsiung, Taiwan
| | - Ing-Jou Chen
- Department of Nuclear Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Pao-Yeh Wang
- Brain Research Center, School of Medicine, National Defense Medical Center Taipei, Taiwan
| | - Ching-Po Lin
- Institute of Neuroscience, National Chaio Tung University, Taipei, Taiwan
| | - Hey-Jen Tsay
- Institute of Neuroscience, National Chaio Tung University, Taipei, Taiwan
| | - Skye Hsin-Hsien Yeh
- Brain Research Center, National Chaio Tung University, Taipei, Taiwan
- Brain Research Center, School of Medicine, National Defense Medical Center Taipei, Taiwan
| |
Collapse
|
17
|
Almotiri A, Abdelfattah A, Storch E, Stemmler MP, Brabletz S, Brabletz T, Rodrigues NP. Zeb1 maintains long-term adult hematopoietic stem cell function and extramedullary hematopoiesis. Exp Hematol 2024; 134:104177. [PMID: 38336135 DOI: 10.1016/j.exphem.2024.104177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
Emerging evidence implicates the epithelial-mesenchymal transition transcription factor Zeb1 as a critical regulator of hematopoietic stem cell (HSC) differentiation. Whether Zeb1 regulates long-term maintenance of HSC function remains an open question. Using an inducible Mx-1-Cre mouse model that deletes conditional Zeb1 alleles in the adult hematopoietic system, we found that mice engineered to be deficient in Zeb1 for 32 weeks displayed expanded immunophenotypically defined adult HSCs and multipotent progenitors associated with increased abundance of lineage-biased/balanced HSC subsets and augmented cell survival characteristics. During hematopoietic differentiation, persistent Zeb1 loss increased B cells in the bone marrow and spleen and decreased monocyte generation in the peripheral blood. In competitive transplantation experiments, we found that HSCs from adult mice with long-term Zeb1 deletion displayed a cell autonomous defect in multilineage differentiation capacity. Long-term Zeb1 loss perturbed extramedullary hematopoiesis characterized by increased splenic weight and a paradoxical reduction in splenic cellularity that was accompanied by HSC exhaustion, lineage-specific defects, and an accumulation of aberrant, preleukemic like c-kit+CD16/32+ progenitors. Loss of Zeb1 for up to 42 weeks can lead to progressive splenomegaly and an accumulation of Gr-1+Mac-1+ cells, further supporting the notion that long-term expression of Zeb1 suppresses preleukemic activity. Thus, sustained Zeb1 deletion disrupts HSC functionality in vivo and impairs regulation of extramedullary hematopoiesis with potential implications for tumor suppressor functions of Zeb1 in myeloid neoplasms.
Collapse
Affiliation(s)
- Alhomidi Almotiri
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Shaqra University, Dawadmi, Saudi Arabia; European Cancer Stem Cell Research Institute, Cardiff University, School of Biosciences, Cardiff, UK
| | - Ali Abdelfattah
- European Cancer Stem Cell Research Institute, Cardiff University, School of Biosciences, Cardiff, UK; Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Elis Storch
- European Cancer Stem Cell Research Institute, Cardiff University, School of Biosciences, Cardiff, UK
| | - Marc P Stemmler
- Department of Experimental Medicine, Nikolaus-Fiebiger-Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Brabletz
- Department of Experimental Medicine, Nikolaus-Fiebiger-Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine, Nikolaus-Fiebiger-Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Neil P Rodrigues
- European Cancer Stem Cell Research Institute, Cardiff University, School of Biosciences, Cardiff, UK.
| |
Collapse
|
18
|
Tóth F, Moftakhar Z, Sotgia F, Lisanti MP. In Vitro Investigation of Therapy-Induced Senescence and Senescence Escape in Breast Cancer Cells Using Novel Flow Cytometry-Based Methods. Cells 2024; 13:841. [PMID: 38786063 PMCID: PMC11120107 DOI: 10.3390/cells13100841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Although cellular senescence was originally defined as an irreversible form of cell cycle arrest, in therapy-induced senescence models, the emergence of proliferative senescence-escaped cancer cells has been reported by several groups, challenging the definition of senescence. Indeed, senescence-escaped cancer cells may contribute to resistance to cancer treatment. Here, to study senescence escape and isolate senescence-escaped cells, we developed novel flow cytometry-based methods using the proliferation marker Ki-67 and CellTrace CFSE live-staining. We investigated the role of a novel senescence marker (DPP4/CD26) and a senolytic drug (azithromycin) on the senescence-escaping ability of MCF-7 and MDA-MB-231 breast cancer cells. Our results show that the expression of DPP4/CD26 is significantly increased in both senescent MCF-7 and MDA-MB-231 cells. While not essential for senescence induction, DPP4/CD26 contributed to promoting senescence escape in MCF-7 cells but not in MDA-MB-231 cells. Our results also confirmed the potential senolytic effect of azithromycin in senescent cancer cells. Importantly, the combination of azithromycin and a DPP4 inhibitor (sitagliptin) demonstrated a synergistic effect in senescent MCF-7 cells and reduced the number of senescence-escaped cells. Although further research is needed, our results and novel methods could contribute to the investigation of the mechanisms of senescence escape and the identification of potential therapeutic targets. Indeed, DPP4/CD26 could be a promising marker and a novel target to potentially decrease senescence escape in cancer.
Collapse
Affiliation(s)
- Fanni Tóth
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
- The CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Wien, Vienna, Austria
| | - Zahra Moftakhar
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| | - Federica Sotgia
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| | - Michael P. Lisanti
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| |
Collapse
|
19
|
Yang J, Zou S, Qiu Z, Lai M, Long Q, Chen H, Lai PL, Zhang S, Rao Z, Xie X, Gong Y, Liu A, Li M, Bai X. Mecp2 fine-tunes quiescence exit by targeting nuclear receptors. eLife 2024; 12:RP89912. [PMID: 38747706 PMCID: PMC11095939 DOI: 10.7554/elife.89912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Abstract
Quiescence (G0) maintenance and exit are crucial for tissue homeostasis and regeneration in mammals. Here, we show that methyl-CpG binding protein 2 (Mecp2) expression is cell cycle-dependent and negatively regulates quiescence exit in cultured cells and in an injury-induced liver regeneration mouse model. Specifically, acute reduction of Mecp2 is required for efficient quiescence exit as deletion of Mecp2 accelerates, while overexpression of Mecp2 delays quiescence exit, and forced expression of Mecp2 after Mecp2 conditional knockout rescues cell cycle reentry. The E3 ligase Nedd4 mediates the ubiquitination and degradation of Mecp2, and thus facilitates quiescence exit. A genome-wide study uncovered the dual role of Mecp2 in preventing quiescence exit by transcriptionally activating metabolic genes while repressing proliferation-associated genes. Particularly disruption of two nuclear receptors, Rara or Nr1h3, accelerates quiescence exit, mimicking the Mecp2 depletion phenotype. Our studies unravel a previously unrecognized role for Mecp2 as an essential regulator of quiescence exit and tissue regeneration.
Collapse
Affiliation(s)
- Jun Yang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Shitian Zou
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zeyou Qiu
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Mingqiang Lai
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Qing Long
- Department of Biochemistry, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Huan Chen
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Ping lin Lai
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Sheng Zhang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zhi Rao
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Xiaoling Xie
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yan Gong
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Anling Liu
- Department of Biochemistry, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Mangmang Li
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
20
|
Gayan S, Teli A, Sonawane A, Dey T. Impact of Chemotherapeutic Stress Depends on The Nature of Breast Cancer Spheroid and Induce Behavioral Plasticity to Resistant Population. Adv Biol (Weinh) 2024; 8:e2300271. [PMID: 38063815 DOI: 10.1002/adbi.202300271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 11/20/2023] [Indexed: 04/15/2024]
Abstract
Cellular or tumor dormancy, identified recently as one of the main reasons behind post-therapy recurrence, can be caused by diverse reasons. Chemotherapy has recently been recognized as one of such reasons. However, in-depth studies of chemotherapy-induced dormancy are lacking due to the absence of an in vitro human-relevant model tailor-made for such a scenario. This report utilized multicellular breast cancer spheroid to create a primary platform for establishing a chemotherapy-induced dormancy model. It is observed that extreme chemotherapeutic stress affects invasive and non-invasive spheroids differently. Non-invasive spheroids exhibit more resilience and maintain viability and migrational ability, while invasive spheroids display heightened susceptibility and improved tumorigenic capacity. Heterogenous spheroids exhibit increased tumorigenic capacity while show minimal survival ability. Further probing of chemotherapeutically dormant spheroids is needed to understand the molecular mechanism and identify dormancy-related markers to achieve therapeutic success in the future.
Collapse
Affiliation(s)
- Sukanya Gayan
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Abhishek Teli
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Akshay Sonawane
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Tuli Dey
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| |
Collapse
|
21
|
Zhang Z, Li X, Ma L, Wang S, Zhang J, Zhou Y, Guo X, Niu Q. LNC000152 Mediates Aluminum-Induced Proliferation of Reactive Astrocytes. ACS OMEGA 2024; 9:11958-11968. [PMID: 38496998 PMCID: PMC10938322 DOI: 10.1021/acsomega.3c09702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 03/19/2024]
Abstract
Aluminum is a metal element with significant neurotoxicity, and there is a substantial correlation between aluminum exposure and cognitive dysfunction. Glial fibrillary acidic protein (GFAP) is widely used as a marker of reactive astrocyte proliferation in response to pathological injury of the central nervous system. Studies of various neurodegenerative diseases have confirmed that the expression changes in GFAP are associated with nerve injury. We investigated the role of LNC000152 in the aluminum-induced reactive proliferation of astrocytes. By establishing two aluminum-exposed cell models of rat primary astrocytes and CTX-TNA2 cell lines, we examined the expression of LNC000152 and GFAP and detected cell proliferation with EdU and cell cycle changes with flow cytometry. The role of aluminum in promoting glial cell proliferation was verified; the expression levels of LNC000152 and GFAP increased with the concentration of aluminum exposure. Intervention of LNC000152 expression by siRNA technology revealed that LNC000152 affected glial cell responsive proliferation by influencing GFAP expression. These results suggest that LNC000152 plays a role in the reactive proliferation of astrocytes induced by aluminum.
Collapse
Affiliation(s)
- Zhuoran Zhang
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaoyan Li
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Limin Ma
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Shanshan Wang
- Section
of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jingsi Zhang
- Section
of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yue Zhou
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xin Guo
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Qiao Niu
- Department
of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| |
Collapse
|
22
|
Abdulrahman FG, Abulkhair HS, Saeed HSE, El-Dydamony NM, Husseiny EM. Design, synthesis, and mechanistic insight of novel imidazolones as potential EGFR inhibitors and apoptosis inducers. Bioorg Chem 2024; 144:107105. [PMID: 38219482 DOI: 10.1016/j.bioorg.2024.107105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
As regards to the structural analysis and optimization of diverse potential EGFR inhibitors, two series of imidazolyl-2-cyanoprop-2-enimidothioates and ethyl imidazolylthiomethylacrylates were designed and constructed as potential EGFR suppressors. The cytotoxic effect of the prepared derivatives was assessed toward hepatic, breast, and prostate cancerous cells (Hep-G2, MCF-7, and PC-3). Three derivatives 3d, 3e, and 3f presented potent antiproliferative activity and selectivity against the examined tumor cells showing IC50 values at low micromolar levels. Hence, successive biological assays were applied to determine the probable mechanism of action of the new compounds. They exhibited significant EGFR suppression with an IC50 range of 0.137-0.507 µM. The most effective EGFR inhibitor 3f arrested the MCF-7 cell cycle at the S phase by inducing the apoptotic pathway that was confirmed via increasing the expression of Caspases 8, 9, and Bax, which are associated with Bcl-2 decline. Additionally, molecular docking displayed a distinctive interaction between 3f and EGFR binding pocket. Overall, this work introduces some novel imidazolyl-2-cyanoprop-2-enimidothioates and ethyl imidazolylthiomethylacrylates as potential cytotoxic and EGFR inhibitors that deserve further research in tumor therapy.
Collapse
Affiliation(s)
- Fatma G Abdulrahman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11884, Cairo, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, International Coastal Road, New Damietta 34518, Egypt.
| | - Hoda S El Saeed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Nehad M El-Dydamony
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6(th) of October City, Egypt
| | - Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt.
| |
Collapse
|
23
|
Sireesha K, Samundeshwari EL, Surekha K, Chandrasekhar C, Sarma PVGK. In vitro generation of epidermal keratinocytes from human CD34-positive hematopoietic stem cells. In Vitro Cell Dev Biol Anim 2024; 60:236-248. [PMID: 38502372 DOI: 10.1007/s11626-024-00862-x] [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: 01/03/2024] [Accepted: 02/05/2024] [Indexed: 03/21/2024]
Abstract
The epidermis is largely composed of keratinocytes (KCs), and the proliferation and differentiation of KCs from the stratum basale to the stratum corneum is the cellular hierarchy present in the epidermis. In this study, we explore the differentiation abilities of human hematopoietic stem cells (HSCs) into KCs. Cultured HSCs positive for CD34, CD45, and CD133 with prominent telomerase activity were induced with keratinocyte differentiation medium (KDM), which is composed of bovine pituitary extract (BPE), epidermal growth factor (EGF), insulin, hydrocortisone, epinephrine, transferrin, calcium chloride (CaCl2), bone morphogenetic protein 4 (BMP4), and retinoic acid (RA). Differentiation was monitored through the expression of cytokeratin markers K5 (keratin 5), K14 (keratin 14), K10 (keratin 10), K1 (keratin 1), transglutaminase 1 (TGM1), involucrin (IVL), and filaggrin (FLG) on day 0 (D0), day 6 (D6), day 11 (D11), day 18 (D18), day 24 (D24), and day 30 (D30) using immunocytochemistry, fluorescence microscopy, flow cytometry, qPCR, and Western blotting. The results revealed the expression of K5 and K14 genes in D6 cells (early keratinocytes), K10 and K1 genes in D11-D18 cells (mature keratinocytes) with active telomerase enzyme, and FLG, IVL, and TGM1 in D18-D24 cells (terminal keratinocytes), and by D30, the KCs were completely enucleated similar to cornified matrix. This method of differentiation of HSCs to KCs explains the cellular order exists in the normal epidermis and opens the possibility of exploring the use of human HSCs in the epidermal differentiation.
Collapse
Affiliation(s)
- Kodavala Sireesha
- Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences and University, Tirupati, 517507, Andhra Pradesh, India
| | | | - Kattaru Surekha
- Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences and University, Tirupati, 517507, Andhra Pradesh, India
| | - Chodimella Chandrasekhar
- Department of Hematology, Sri Venkateswara Institute of Medical Sciences, Tirupati, 517507, Andhra Pradesh, India
| | | |
Collapse
|
24
|
Lazarski CA, Hanley PJ. Review of flow cytometry as a tool for cell and gene therapy. Cytotherapy 2024; 26:103-112. [PMID: 37943204 PMCID: PMC10872958 DOI: 10.1016/j.jcyt.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Quality control testing and analytics are critical for the development and manufacture of cell and gene therapies, and flow cytometry is a key quality control and analytical assay that is used extensively. However, the technical scope of characterization assays and safety assays must keep apace as the breadth of cell therapy products continues to expand beyond hematopoietic stem cell products into producing novel adoptive immune therapies and gene therapy products. Flow cytometry services are uniquely positioned to support the evolving needs of cell therapy facilities, as access to flow cytometers, new antibody clones and improved fluorochrome reagents becomes more egalitarian. This report will outline the features, logistics, limitations and the current state of flow cytometry within the context of cellular therapy.
Collapse
Affiliation(s)
- Christopher A Lazarski
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| | - Patrick J Hanley
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| |
Collapse
|
25
|
Khamis AA, Ali EMM, Salim EI, El-Moneim MAA. Synergistic effects of bee venom, hesperidin, and piperine with tamoxifen on apoptotic and angiogenesis biomarker molecules against xerographic MCF-7 injected rats. Sci Rep 2024; 14:1510. [PMID: 38233443 PMCID: PMC10794414 DOI: 10.1038/s41598-023-50729-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 12/23/2023] [Indexed: 01/19/2024] Open
Abstract
Breast cancer ranks as the second leading most significant of mortality for women. Studies have demonstrated the potential benefits of natural compounds in cancer treatment and prevention, either in isolation or in conjunction with chemotherapy. In order to improve Tamoxifen's therapeutic efficacy in in-vivo studies, our research sought to determine the effects of hesperidin, piperine, and bee venom as natural compounds, as well as their combination effect with or without Tamoxifen. First, 132 female albino rats were equally divided into six groups and five subgroups, and breast cancer was induced in the selected groups by xenografting of MCF7 cells. Second, the effect of single and best ratio combinations treatment from previous in vitro studies were selected. Next, tumorous mammary glands were collected for apoptotic and antiapoptotic biomarkers and cell cycle analysis. Single or combined natural products with or without Tamoxifen revealed a significant up-regulation in apoptotic genes Bax and Casp3 and a downregulation of antiapoptotic and angiogenesis genes Bcl-2 and VEGF genes. We found that cell cycle arrest in the G0/G1 phase was exclusively caused by Tamoxifen and/ or hesperidin. However, the cell cycle arrest in the G2/M phase is a result of the combination of piperine and bee venom, with or without Tamoxifen by using the flow cytometric technique. Our research concludes that bee venom, hesperidin, and piperine can synergistically enhance to increase Tamoxifen's efficiency in the management of breast cancer.
Collapse
Affiliation(s)
- Abeer A Khamis
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Ehab M M Ali
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Elsayed I Salim
- Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed A Abd El-Moneim
- Biochemistry Department, Faculty of Dentistry, Sinai University, Al-Arish, North Sinai, Egypt
| |
Collapse
|
26
|
Abdulrahman FG, Abulkhair HS, Zidan RA, Alwakeel AI, Al-Karmalawy AA, Husseiny EM. Novel benzochromenes: design, synthesis, cytotoxicity, molecular docking and mechanistic investigations. Future Med Chem 2024; 16:105-123. [PMID: 38226455 DOI: 10.4155/fmc-2023-0198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/29/2023] [Indexed: 01/17/2024] Open
Abstract
Aim: A novel series of fused benzochromenes with expected cytotoxicity and HIF-1α inhibition was identified. Materials & methods: A bioisosterism-aided approach was applied to design new benzochromenes and assess their cytotoxicity against three cancer cell lines. The probable mechanistic effect and the in silico docking and pharmacokinetic profiles of the most effective derivatives were evaluated. Results: Compounds 3, 4, 5, 8 and 11 showed potent antiproliferative activity and excellent selectivity. Compound 8 showed significant HIF-1α inhibition with an IC50 value of 3.372 μM. It also enhanced apoptosis and arrested the HepG2 cell cycle at both the G0/G1 and S stages. Conclusion: Compound 8 was identified as a new potential anticancer candidate.
Collapse
Affiliation(s)
- Fatma G Abdulrahman
- Pharmaceutical Organic Chemistry Department, Al-Azhar University, Nasr City, 11754, Cairo, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Al-Azhar University, Nasr City, 11884, Cairo, Egypt
- Pharmaceutical Chemistry Department, Horus University-Egypt, International Coastal Road, New Damietta, 34518, Egypt
| | - Riham A Zidan
- Department of Biochemistry, Al-Azhar University, Cairo, Egypt
| | - Asmaa I Alwakeel
- Department of Pharmacology & Toxicology, Al Azhar University, Cairo, Egypt
| | - Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Horus University-Egypt, International Coastal Road, New Damietta, 34518, Egypt
- Pharmaceutical Chemistry Department, Ahram Canadian University, Giza, 12566, Egypt
| | - Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Al-Azhar University, Nasr City, 11754, Cairo, Egypt
| |
Collapse
|
27
|
Alzeer RM, Al-Hadyan KS, Al-Harbi NM, Bin Judia SS, Almeer RS, Alsbeih GA. Cytotoxicity and Radiosensitizing Potentials of Pilosulin-3, a Recombinant Ant Venom, in Breast Cancer Cells. Toxins (Basel) 2023; 15:701. [PMID: 38133205 PMCID: PMC10747674 DOI: 10.3390/toxins15120701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Venom peptides are promising agents in the development of unconventional anticancer therapeutic agents. This study explored the potential of Pilosulin-3, a recombinant peptide from the venom of the Australian jack jumper ant "Myrmecia pilosula", as a cytotoxic and radiosensitizing agent in MCF-7 and MDA-MB-231 breast cancer (BC) cell lines. Pilosulin-3's cytotoxicity was evaluated across a wide range of concentrations using a proliferation assay. Cell cycle progression and apoptosis were examined at the inhibitory concentration 25% (IC25) and IC50 of Pilosulin-3, both with and without a 4Gy X-ray irradiation dose. Radiosensitivity was assessed at IC25 using the clonogenic survival assay. The study revealed that Pilosulin-3 exerted a concentration-dependent cytotoxic effect, with IC25 and IC50 values of 0.01 and 0.5 µM, respectively. In silico screening indicated high selectivity of Pilosulin-3 peptide, which was predicted to be the most likely anticancer agent (PROB = 0.997) with low hemolytic activity (PROP = 0.176). Although Pilosulin-3 exhibited a significant (p < 0.05) G2/M cell cycle arrest in combination with radiation, there was no discernible effect on apoptosis induction or cell survival following irradiation. In conclusion, Pilosulin-3 proved to be cytotoxic to BC cells and induced a cytostatic effect (G2/M arrest) when combined with radiation. However, it did not enhance the efficacy of cell killing by irradiation. While it holds potential as a cytotoxic agent in breast cancer treatment, its application as a radiosensitizer does not find support in these results.
Collapse
Affiliation(s)
- Reema M. Alzeer
- Department of Zoology, College of Science, King Saud University, Riyadh 11541, Saudi Arabia
| | - Khaled S. Al-Hadyan
- Biomedical Physics Department, Radiation Biology Section, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| | - Najla M. Al-Harbi
- Biomedical Physics Department, Radiation Biology Section, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| | - Sara S. Bin Judia
- Biomedical Physics Department, Radiation Biology Section, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| | - Rafa S. Almeer
- Department of Zoology, College of Science, King Saud University, Riyadh 11541, Saudi Arabia
| | - Ghazi A. Alsbeih
- Biomedical Physics Department, Radiation Biology Section, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
28
|
Jayaraman S, Raj Natarajan S, Ponnusamy B, Veeraraghavan VP, Jasmine S. Unlocking the potential of beta sitosterol: Augmenting the suppression of oral cancer cells through extrinsic and intrinsic signalling mechanisms. Saudi Dent J 2023; 35:1007-1013. [PMID: 38107042 PMCID: PMC10724352 DOI: 10.1016/j.sdentj.2023.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 12/19/2023] Open
Abstract
The global increase in the prevalence of oral neoplasms and related deaths can be attributed to social development and lifestyle factors, leading to poor prognosis and a lack of early clinical detection. Oral cancer ranks ranked sixth mostly diagnosed cancer and is a leading cause of cancer-related deaths. In light of these circumstances, our objective was to assess the potential of β-sitosterol, a naturally occurring herbal compound, as an anticancer agent against KB cells, a representative cell line for oral cancer. Our study primarily focused on evaluating the cytotoxic effect and mRNA expression of apoptotic proteins by β-sitosterol on KB cells. The results demonstrated a remarkable cytotoxic effect, leading to cell death. Further investigation using flow cytometric analysis revealed that this cell death was mediated through the initiation of the apoptotic signalling by β-sitosterol. The use of the bioinformatic tool, STITCH, supported our study by predicting drug-protein interactions and suggesting that β-sitosterol may play a significant role in targeting apoptotic pathways. Additionally, docking results were employed to validate the findings demonstrating high binding affinity of β-sitosterol with apoptotic-mediated signalling targets. To gain deeper insights into the molecular insights, we measured mRNA levels for BAX, BCL-2, MCL-1, P53, P21, MDM2, caspase3, and caspase9. Based on our comprehensive findings, our study concludes that β-sitosterol holds significant therapeutic potential against oral cancer cells. These results strongly suggest that this herbal compound should be further explored as a potential treatment option for oral cancer for clinical trial.
Collapse
Affiliation(s)
- Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Sathan Raj Natarajan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Bhuvaneswari Ponnusamy
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Sharmila Jasmine
- Department of Oral Maxillofacial Surgery, Rajas Dental College and Hospital, Kavalkinaru, Tirunelveli 627105, Tamil Nadu, India
| |
Collapse
|
29
|
Husseiny EM, Abulkhair HS, Saleh A, Altwaijry N, Zidan RA, Abdulrahman FG. Molecular overlay-guided design of new CDK2 inhibitor thiazepinopurines: Synthesis, anticancer, and mechanistic investigations. Bioorg Chem 2023; 140:106789. [PMID: 37611530 DOI: 10.1016/j.bioorg.2023.106789] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023]
Abstract
Adopting the molecular overlay approach, three novel sets of thiazepinopurines with expected cytotoxicity and CDK2 inhibition potential were designed and synthesized. This was accomplished through the heteroannelation of purines, for the first time, with thiazepine. The obtained thiazepinopurines derivatives were assessed for their cytotoxicity toward tumor cells of three different types, HepG2, MCF-7, and PC-3 as well as one normal cell (WI38). Among the studied compounds, 3b and 3c exhibited significant antiproliferative activity against tumor cells presenting IC50 range of 5.52-17.09 µM in comparison with Roscovitine (9.32-13.82 µM). Additionally, both compounds displayed superior selectivity indices (SI = 3.00-7.15) toward tested cancer cells. The 4-chlorophenyl analog 3b has shown the best selectivity index, and hence it has been subjected to additional investigations to determine its proper mechanistic effect. Accordingly, the CDK2 inhibition potential, apoptosis induction, and cell cycle analysis of MCF-7 were evaluated. Results revealed that this analog displayed a potent CDK2 inhibition potential with an IC50 value of 0.219 µM. Findings also showed that 3b was thought to arrest MCF-7 cell cycle at S phase together with apoptosis induction by the increased expression of Bax, Caspase-8, and -9 markers with a concomitant decrease in Bcl-2 expression. Besides, the probable interaction of 3b with CDK2 binding pocket was investigated by molecular docking.
Collapse
Affiliation(s)
- Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11884, Cairo, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, International Coastal Road, New Damietta 34518, Egypt
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Najla Altwaijry
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Riham A Zidan
- Department of Biochemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Fatma G Abdulrahman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| |
Collapse
|
30
|
Swathi K, Sumathi S, Somit K, Shubashini SK. Reverse pharmacology approach to validate the diabetic wound-healing activity of Jatyadi thailam formulations in vitro on diabetic mimic environment. Arch Physiol Biochem 2023:1-14. [PMID: 37897224 DOI: 10.1080/13813455.2023.2264536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023]
Abstract
Objective: Jatyadi thailam, an Ayurvedic preparation, is renowned for its efficacy in diabetic wound healing and inflammation. This study aimed to validate and compare the diabetic wound-healing potential of two Jatyadi thailam formulations - Ayurvedic formulary of India Jatyadi thailam (JT-AFI) and Yogagrantha formulation of Jatyadi thailam (JT-YG), in a diabetic environment using L929 fibroblast cells in vitro. Methodology: The effects on cell survival, proliferation, migration, angiogenesis, cell cycle progression, apoptosis, ROS generation, and mitochondrial function were evaluated.Results: The formulations promoted cell proliferation, migration, angiogenesis, while also regulating cell cycle and apoptosis. They effectively suppressed ROS generation and modulated mitochondrial function. JT-AFI exhibited superior efficacy in accelerating diabetic wound healing compared to JT-YG.Conclusion: These findings provide substantial support for the mechanistic role of Jatyadi thailam in diabetic wound healing.
Collapse
Affiliation(s)
- Kandasamy Swathi
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, India
| | - Sundaravadivelu Sumathi
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, India
| | | | - Sripathi K Shubashini
- Department of Chemistry, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, India
| |
Collapse
|
31
|
Gordon T, Hendin N, Wurtzel O. Methods for cell isolation and analysis of the highly regenerative tunicate Polycarpa mytiligera. Front Cell Dev Biol 2023; 11:1274826. [PMID: 37886396 PMCID: PMC10598751 DOI: 10.3389/fcell.2023.1274826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Background: Polycarpa mytiligera is the only molecularly characterized solitary ascidian capable of regenerating all organs and tissue types. The cellular basis for regeneration in P. mytiligera is largely unknown, and methods for isolating live cells from this species for functional analyses are unavailable. Results: Here, we developed a method for isolating live cells from P. mytiligera, overcoming major experimental challenges, including the dissociation of its thick body wall and native cellular autofluorescence. We demonstrated the applicability of our approach for tissue dissociation and cell analysis using three flow cytometry platforms, and by using broadly used non-species-specific cell labeling reagents. In addition to live cell isolation, proof-of-concept experiments showed that this approach was compatible with gene expression analysis of RNA extracted from the isolated cells, and with ex vivo analysis of phagocytosis. Conclusion: We presented efficient methods for cell purification from a highly regenerative ascidian, which could be transferable to diversity of non-model marine organisms. The ability to purify live cells will promote future studies of cell function in P. mytiligera regeneration.
Collapse
Affiliation(s)
- Tal Gordon
- The School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Noam Hendin
- The School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Omri Wurtzel
- The School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
32
|
TAMURA N, HEIDARI N, FARAGHER RG, SMITH RK, DUDHIA J. Effects of resveratrol and its analogues on the cell cycle of equine mesenchymal stem/stromal cells. J Equine Sci 2023; 34:67-72. [PMID: 37781569 PMCID: PMC10534064 DOI: 10.1294/jes.34.67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/28/2023] [Indexed: 10/03/2023] Open
Abstract
Resveratrol (RSV; trans-3,5,4'-trihydroxystilbene) strongly activates sirtuin 1, and it and its analogue V29 enhance the proliferation of mesenchymal stem/stromal cells (MSCs).Although culture medium containing 5-azacytydine and RSV inhibits senescence of adipose tissue-derived MSCs isolated from horses with metabolic syndrome, few studies have reported the effects of RSV on equine bone marrow-derived MSCs (eBMMSCs) isolated from horses without metabolic syndrome. The aim of this study was to investigate the effects of RSV and V29 on the cell cycle of eBMMSCs. Following treatment with 5 µM RSV or 10 µM V29, the cell proliferation capacity of eBMMSCs derived from seven horses was evaluated by EdU (5-ethynyl-2'-deoxyuridine) and Ki-67 antibody assays. Brightfield images of cells and immunofluorescent images of EdU, Ki-67, and DAPI staining were recorded by fluorescence microscopy, and the number of cells positive for each was quantified and compared by Friedman's test at P<0.05. The growth fraction of eBMMSCs was significantly increased by RSV and V29 as measured by the EdU assay (control 28.1% ± 13.8%, V29 31.8% ± 14.6%, RSV 32.0% ± 10.8%; mean ± SD; P<0.05) but not as measured by the Ki-67 antibody assay (control 27.0% ± 11.2%, V29 27.4% ± 10.8%, RSV 27.7% ± 6.8%). RSV and V29 promoted progression of the cell cycle of eBMMSCs into the S phase and may be useful for eBMMSC expansion.
Collapse
Affiliation(s)
- Norihisa TAMURA
- Clinical Veterinary Medicine Division, Equine
Research Institute, Japan Racing Association, Tochigi 329-0412,
Japan
- Department of Clinical Sciences and Services,
Royal Veterinary College, University of London, Hatfield AL9 7TA, UK
| | - Neda HEIDARI
- Department of Clinical Sciences and Services,
Royal Veterinary College, University of London, Hatfield AL9 7TA, UK
| | - Richard G.A. FARAGHER
- School of Pharmacy and Biomolecular Sciences,
University of Brighton, Brighton BN2 4GJ, UK
| | - Roger K.W. SMITH
- Department of Clinical Sciences and Services,
Royal Veterinary College, University of London, Hatfield AL9 7TA, UK
| | - Jayesh DUDHIA
- Department of Clinical Sciences and Services,
Royal Veterinary College, University of London, Hatfield AL9 7TA, UK
| |
Collapse
|
33
|
Klapp V, Bloy N, Jiménez-Cortegana C, Buqué A, Petroni G. Flow cytometry-assisted quantification of cell cycle arrest in cancer cells treated with CDK4/6 inhibitors. Methods Cell Biol 2023; 181:197-212. [PMID: 38302240 DOI: 10.1016/bs.mcb.2023.02.018] [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: 02/03/2024]
Abstract
Cyclin-dependent kinase 4 (CDK4) and CDK6 inhibitors (i.e., palbociclib, abemaciclib, and ribociclib) are well known for their capacity to mediate cytostatic effects by promoting cell cycle arrest in the G1 phase, thus inhibiting cancer cell proliferation. Cytostatic effects induced by CDK4/6 inhibitors can be transient or lead to a permanent state of cell cycle arrest, commonly defined as cellular senescence. Induction of senescence is often associated to metabolic modifications and to the acquisition of a senescence-associated secretory phenotype (SASP) by cancer cells, which in turn can promote or limit antitumor immunity (and thus the efficacy of CDK4/6 inhibitors) depending on SASP components. Thus, although accumulating evidence suggests that anti-cancer effects of CDK4/6 inhibitors also depend on the promotion of antitumor immune responses, assessing cell cycle arrest and progression in cells treated with palbociclib remains a key approach for investigating the efficacy of CDK4/6 inhibitors. Here, we describe a method to assess cell cycle distribution simultaneously with active DNA replication by flow cytometry in cultured hormone receptor-positive breast cancer MCF7 cells.
Collapse
Affiliation(s)
- Vanessa Klapp
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY, United States; Tumor Stroma Interactions, Department of Cancer Research, Luxembourg, Institute of Health, Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Norma Bloy
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY, United States
| | - Carlos Jiménez-Cortegana
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY, United States
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY, United States.
| | - Giulia Petroni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
34
|
Sebastian S, Hoffmann MK, Howard D, Young C, Washington J, Unterweger H, Alexiou C, Turnbull T, D’Andrea R, Hoffmann P, Kempson I. Kinetic Effects of Transferrin-Conjugated Gold Nanoparticles on the Antioxidant Glutathione-Thioredoxin Pathway. Antioxidants (Basel) 2023; 12:1617. [PMID: 37627612 PMCID: PMC10451790 DOI: 10.3390/antiox12081617] [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: 06/01/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Nanoparticle-based therapeutics are being clinically translated for treating cancer. Even when thought to be biocompatible, nanoparticles are being increasingly identified as altering cell regulation and homeostasis. Antioxidant pathways are important for maintaining cell redox homeostasis and play important roles by maintaining ROS levels within tolerable ranges. Here, we sought to understand how a model of a relatively inert nanoparticle without any therapeutic agent itself could antagonize a cancer cell lines' antioxidant mechanism. A label-free protein expression approach was used to assess the glutathione-thioredoxin antioxidative pathway in a prostate cancer cell line (PC-3) after exposure to gold nanoparticles conjugated with a targeting moiety (transferrin). The impact of the nanoparticles was also corroborated through morphological analysis with TEM and classification of pro-apoptotic cells by way of the sub-G0/G1 population via the cell cycle and annexin V apoptosis assay. After a two-hour exposure to nanoparticles, major proteins associated with the glutathione-thioredoxin antioxidant pathway were downregulated. However, this response was acute, and in terms of protein expression, cells quickly recovered within 24 h once nanoparticle exposure ceased. The impact on PRDX-family proteins appears as the most influential factor in how these nanoparticles induced an oxidative stress response in the PC-3 cells. An apparent adaptive response was observed if exposure to nanoparticles continued. Acute exposure was observed to have a detrimental effect on cell viability compared to continuously exposed cells. Nanoparticle effects on cell regulation likely provide a compounding therapeutic advantage under some circumstances, in addition to the action of any cytotoxic agents; however, any therapeutic advantage offered by nanoparticles themselves with regard to vulnerabilities specific to the glutathione-thioredoxin antioxidative pathway is highly temporal.
Collapse
Affiliation(s)
- Sonia Sebastian
- Future Industries Institute, University of South Australia, Adelaide, SA 5095, Australia; (S.S.); (D.H.); (T.T.)
- Clinical Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (M.K.H.); (C.Y.); (P.H.)
| | - Manuela Klingler Hoffmann
- Clinical Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (M.K.H.); (C.Y.); (P.H.)
- Mass Spectrometry & Proteomics Group, Clinical Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Douglas Howard
- Future Industries Institute, University of South Australia, Adelaide, SA 5095, Australia; (S.S.); (D.H.); (T.T.)
| | - Clifford Young
- Clinical Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (M.K.H.); (C.Y.); (P.H.)
- Mass Spectrometry & Proteomics Group, Clinical Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Jenni Washington
- Clinical Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (M.K.H.); (C.Y.); (P.H.)
- Mass Spectrometry & Proteomics Group, Clinical Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Harald Unterweger
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (H.U.); (C.A.)
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (H.U.); (C.A.)
| | - Tyron Turnbull
- Future Industries Institute, University of South Australia, Adelaide, SA 5095, Australia; (S.S.); (D.H.); (T.T.)
| | - Richard D’Andrea
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia;
| | - Peter Hoffmann
- Clinical Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (M.K.H.); (C.Y.); (P.H.)
- Mass Spectrometry & Proteomics Group, Clinical Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Ivan Kempson
- Future Industries Institute, University of South Australia, Adelaide, SA 5095, Australia; (S.S.); (D.H.); (T.T.)
| |
Collapse
|
35
|
Yadav N, Babu D, Madigubba S, Panigrahi M, Phanithi PB. Tyrphostin A9 attenuates glioblastoma growth by suppressing PYK2/EGFR-ERK signaling pathway. J Neurooncol 2023; 163:675-692. [PMID: 37415005 DOI: 10.1007/s11060-023-04383-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/24/2023] [Indexed: 07/08/2023]
Abstract
PURPOSE Glioblastoma (GBM) is a fatal primary brain tumor with extremely poor clinical outcomes. The anticancer efficiency of tyrosine kinase inhibitors (TKIs) has been shown in GBM and other cancer, with limited therapeutic outcomes. In the current study, we aimed to investigate the clinical impact of active proline-rich tyrosine kinase-2 (PYK2) and epidermal growth factor receptor (EGFR) in GBM and evaluate its druggability by a synthetic TKI-Tyrphostin A9 (TYR A9). METHODS The expression profile of PYK2 and EGFR in astrocytoma biopsies (n = 48) and GBM cell lines were evaluated through quantitative PCR, western blots, and immunohistochemistry. The clinical association of phospho-PYK2 and EGFR was analyzed with various clinicopathological features and the Kaplan-Meier survival curve. The phospho-PYK2 and EGFR druggability and subsequent anticancer efficacy of TYR A9 was evaluated in GBM cell lines and intracranial C6 glioma model. RESULTS Our expression data revealed an increased phospho-PYK2, and EGFR expression aggravates astrocytoma malignancy and is associated with patients' poor survival. The mRNA and protein correlation analysis showed a positive association between phospho-PYK2 and EGFR in GBM tissues. The in-vitro studies demonstrated that TYR A9 reduced GBM cell growth, cell migration, and induced apoptosis by attenuating PYK2/EGFR-ERK signaling. The in-vivo data showed TYR A9 treatment dramatically reduced glioma growth with augmented animal survival by repressing PYK2/EGFR-ERK signaling. CONCLUSION Altogether, this study report that increased phospho-PYK2 and EGFR expression in astrocytoma was associated with poor prognosis. The in-vitro and in-vivo evidence underlined translational implication of TYR A9 by suppressing PYK2/EGFR-ERK modulated signaling pathway. The schematic diagram displayed proof of concept of the current study indicating activated PYK2 either through the Ca2+/Calmodulin-dependent protein kinase II (CAMKII) signaling pathway or autophosphorylation at Tyr402 induces association to the SH2 domain of c-Src that leads to c-Src activation. Activated c-Src in turn activates PYK2 at other tyrosine residues that recruit Grb2/SOS complex and trigger ERK½ activation. Besides, PYK2 interaction with c-Src acts as an upstream of EGFR transactivator that can activate the ERK½ signaling pathway, which induces cell proliferation and cell survival by increasing anti-apoptotic proteins or inhibiting pro-apoptotic proteins. TYR A9 treatment attenuate GBM cell proliferation and migration; and induce GBM cell death by inhibiting PYK2 and EGFR-induced ERK activation.
Collapse
Affiliation(s)
- Neera Yadav
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Deepak Babu
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Sailaja Madigubba
- Department of Laboratory Medicine, Krishna Institute of Medical Sciences, Secunderabad, Telangana, 500 003, India
| | - Manas Panigrahi
- Department of Neurosurgery, Krishna Institute of Medical Sciences, Secunderabad, Telangana, 500 003, India
| | - Prakash Babu Phanithi
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics School of Life Sciences, University of Hyderabad, Room No: F-23/F-71, Hyderabad, Telangana, 500 046, India.
| |
Collapse
|
36
|
Husseiny EM, Abulkhair HS, El-Hddad SS, Osama N, El-Zoghbi MS. Aminopyridone-linked benzimidazoles: a fragment-based drug design for the development of CDK9 inhibitors. Future Med Chem 2023; 15:1213-1232. [PMID: 37584185 DOI: 10.4155/fmc-2023-0139] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023] Open
Abstract
Aim: A fragment-based design and synthesis of three novel series of aminopyridone-linked benzimidazoles as potential anticancer candidates with significant CDK9 inhibition was implemented. Materials & methods: All synthesized compounds were submitted to National Cancer Institute, 60 cell lines and seven-dose cytotoxicity toward three cancer cells. Results: Compounds 2, 4a, 4c, 4d, 6a and 8a exhibited significant cytotoxicity and selectivity with IC50 range of 7.61-57.75 μM. Regarding the mechanism either in vitro or in silico, 4a, 6a and 8a displayed potent CDK9 inhibition with IC50 value of 0.424-8.461 μM. Compound 6a arrested the cell cycle at S phase and induced apoptosis in MCF-7 cells. Conclusion: Compound 6a is a promising CDK9 inhibitor that warrants additional research for cancer treatment.
Collapse
Affiliation(s)
- Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, 11754, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University - Egypt, New Damietta, 34518, Egypt
| | - Sanadelaslam Sa El-Hddad
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Omar Almukhtar University, Al Bayda, 102345, Libya
| | - Nada Osama
- Biochemistry Department, Faculty of Pharmacy, Menoufia University, Shibin Elkom, Menoufia, 32511, Egypt
| | - Mona S El-Zoghbi
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Menoufia University, Shebin El-Koum, 32511, Egypt
| |
Collapse
|
37
|
Zhang W, Yu L, Xu C, Tang T, Cao J, Chen L, Pang X, Ren W. MRPL51 is a downstream target of FOXM1 in promoting the malignant behaviors of lung adenocarcinoma. Oncol Lett 2023; 26:298. [PMID: 37323822 PMCID: PMC10265367 DOI: 10.3892/ol.2023.13884] [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: 11/16/2022] [Accepted: 04/21/2023] [Indexed: 06/17/2023] Open
Abstract
Mitochondrial ribosome protein L51 (MRPL51) is a 39S subunit protein of the mitochondrial ribosome. Its dysregulation may be involved in non-small cell lung cancer. The present study aimed to explore MRPL51 expression in lung adenocarcinoma (LUAD) and normal lung tissues, as well as its regulatory effects on malignant LUAD behaviors. In addition, the role of forkhead box protein M1 (FOXM1) in MRPL51 transcription was studied. Bioinformatics analysis and subsequent in vitro experiments, including western blotting, immunofluorescent staining, Transwell invasion assay, dual-luciferase assay and chromatin immunoprecipitation quantitative PCR were conducted. The results demonstrated that MRPL51 expression was upregulated at both the mRNA and protein levels in LUAD tissues compared with normal lung tissues. Gene Set Enrichment Analysis demonstrated that LUAD tissues with higher MRPL51 expression also had higher expression levels of genes enriched in multiple gene sets, including 'DNA_REPAIR', 'UNFOLDED_PROTEIN_RESPONSE', 'MYC_TARGETS_V1', 'OXIDATIVE_ PHOSPHORYLATION', 'MTORC1_SIGNALING', 'REACTIVE_OXYGEN_SPECIES_PATHWAY', 'MYC_ TARGETS_V2', 'E2F_TARGETS' and 'G2M_ CHECKPOINT'. MRPL51 expression was positively correlated with 'cell cycle', 'DNA damage', 'DNA repair', epithelial-mesenchymal transition ('EMT'), 'invasion' and 'proliferation' of LUAD cells at the single-cell level. Compared to the negative control, MRPL51 knockdown decreased N-cadherin and vimentin expression but increased E-cadherin expression in A549 and Calu-3 cells. MRPL51 knockdown suppressed cell proliferation, induced G1 phase arrest and decreased cell invasion. Patients with LUAD and higher MRPL51 expression had a significantly shorter overall survival (OS). FOXM1 could bind to the MRPL51 gene promoter and activate its transcription. In conclusion, MRPL51 was transcriptionally activated by FOXM1 in LUAD and contributed to the malignant behaviors of tumor cells, including EMT, cell cycle progression and invasion. High MRPL51 expression may be a prognostic biomarker indicating poor OS.
Collapse
Affiliation(s)
- Wenqian Zhang
- Department of Thoracic Surgery, Peking University Shougang Hospital, Beijing 100144, P.R. China
| | - Lei Yu
- Department of Thoracic Surgery, Peking University Shougang Hospital, Beijing 100144, P.R. China
| | - Cong Xu
- Department of Thoracic Surgery, Peking University Shougang Hospital, Beijing 100144, P.R. China
| | - Tian Tang
- Department of Thoracic Surgery, Peking University Shougang Hospital, Beijing 100144, P.R. China
| | - Jianguang Cao
- Department of Thoracic Surgery, Peking University Shougang Hospital, Beijing 100144, P.R. China
| | - Lei Chen
- Department of Thoracic Surgery, Peking University Shougang Hospital, Beijing 100144, P.R. China
| | - Xinya Pang
- Department of Thoracic Surgery, Peking University Shougang Hospital, Beijing 100144, P.R. China
| | - Weihao Ren
- Department of Thoracic Surgery, Peking University Shougang Hospital, Beijing 100144, P.R. China
| |
Collapse
|
38
|
Choroba K, Filipe B, Świtlicka A, Penkala M, Machura B, Bieńko A, Cordeiro S, Baptista PV, Fernandes AR. In Vitro and In Vivo Biological Activities of Dipicolinate Oxovanadium(IV) Complexes. J Med Chem 2023. [PMID: 37311060 DOI: 10.1021/acs.jmedchem.3c00255] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The work is focused on anticancer properties of dipicolinate (dipic)-based vanadium(IV) complexes [VO(dipic)(N∩N)] bearing different diimines (2-(1H-imidazol-2-yl)pyridine, 2-(2-pyridyl)benzimidazole, 1,10-phenanthroline-5,6-dione, 1,10-phenanthroline, and 2,2'-bipyridine), as well as differently 4,7-substituted 1,10-phenanthrolines. The antiproliferative effect of V(IV) systems was analyzed in different tumors (A2780, HCT116, and HCT116-DoxR) and normal (primary human dermal fibroblasts) cell lines, revealing a high cytotoxic effect of [VO(dipic)(N∩N)] with 4,7-dimethoxy-phen (5), 4,7-diphenyl-phen (6), and 1,10-phenanthroline (8) against HCT116-DoxR cells. The cytotoxicity differences between these complexes can be correlated with their different internalization by HCT116-DoxR cells. Worthy of note, these three complexes were found to (i) induce cell death through apoptosis and autophagy pathways, namely, through ROS production; (ii) not to be cytostatic; (iii) to interact with the BSA protein; (iv) do not promote tumor cell migration or a pro-angiogenic capability; (v) show a slight in vivo anti-angiogenic capability, and (vi) do not show in vivo toxicity in a chicken embryo.
Collapse
Affiliation(s)
- Katarzyna Choroba
- University of Silesia, Institute of Chemistry, Szkolna 9, 40-006 Katowice, Poland
| | - Beatriz Filipe
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Anna Świtlicka
- University of Silesia, Institute of Chemistry, Szkolna 9, 40-006 Katowice, Poland
| | - Mateusz Penkala
- University of Silesia, Institute of Chemistry, Szkolna 9, 40-006 Katowice, Poland
| | - Barbara Machura
- University of Silesia, Institute of Chemistry, Szkolna 9, 40-006 Katowice, Poland
| | - Alina Bieńko
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Sandra Cordeiro
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Pedro V Baptista
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Alexandra R Fernandes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| |
Collapse
|
39
|
Caserta S, Genovese C, Cicero N, Gangemi S, Allegra A. The Anti-Cancer Effect of Cinnamon Aqueous Extract: A Focus on Hematological Malignancies. Life (Basel) 2023; 13:life13051176. [PMID: 37240821 DOI: 10.3390/life13051176] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/10/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Cinnamon is an evergreen and tropical plant of the family Lauraceae, growing particularly in Sri Lanka, whose aqueous extract has been tested in different studies to evaluate its possible use as an anti-cancer compound. Both in vitro and in vivo experiments seem to confirm that it acts on various cellular pathways, contributing to down-regulating the activity of molecules that stimulate the proliferation and survival of cells such as the transcription factors NF-KB and AP-1, COX-2, dihydrofolate reductase and pro-angiogenic substances such as VEGF, while up-regulating the function of immune cells against tumors, such as cytotoxic CD8+ T cells. In hematological malignancies, aqueous cinnamon extract has been studied in order to understand if it is possible to count on its help, alone or in combination with traditional drugs such as doxorubicin, to treat patients. The aim of our work is to investigate results from in vitro and in vivo studies about the possible anti-cancer effect of aqueous cinnamon extract in hematological malignancies and the different pathways involved in its action. The possibility of using cinnamon extract in clinical practice is discussed; even if its use could appear very interesting, more studies are necessary to clear the real potentiality of this substance in cancer.
Collapse
Affiliation(s)
- Santino Caserta
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Claudia Genovese
- National Research Council, Institute for Agricultural and Forest Systems in the Mediterranean, Via Empedocle 58, 95128 Catania, Italy
| | - Nicola Cicero
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| |
Collapse
|
40
|
Roškar Z, Dreisinger M, Tič P, Homšak E, Bevc S, Goropevšek A. New Flow Cytometric Methods for Monitoring STAT5 Signaling Reveal Responses to SARS-CoV-2 Antigen-Specific Stimulation in FOXP3+ Regulatory T Cells also in Patients with Advanced Chronic Lymphocytic Leukemia. BIOSENSORS 2023; 13:bios13050539. [PMID: 37232900 DOI: 10.3390/bios13050539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
Increased frequency of CD4+CD25+ regulatory T-cells (Treg) has been associated with disease progression in chronic lymphocytic leukemia (CLL). Flow cytometric methods, which allow for the simultaneous analysis of their specific transcription factor Foxp3 and activated STAT proteins, together with proliferation can help to elucidate the signaling mechanisms driving Treg expansion and suppression of FOXP3- conventional CD4+T-cells (Tcon). Herein, we first report a novel approach in which STAT5 phosphorylation (pSTAT5) and proliferation (BrdU-FITC incorporation) could be analyzed specifically in FOXP3+ and FOXP3- responding cells after CD3/CD28 stimulation. The addition of magnetically purified CD4+CD25+ T-cells from healthy donors to cocultured autologous CD4+CD25- T-cells resulted in suppression of Tcon cell cycle progression accompanied by a decrease in pSTAT5. Next, a method using imaging flow cytometry is presented for the detection of cytokine-dependent pSTAT5 nuclear translocation in FOXP3-expressing cells. Finally, we discuss our experimental data obtained by combining Treg pSTAT5 analysis and antigen-specific stimulation with SARS-CoV-2 antigens. Applying these methods on samples from patients revealed Treg responses to antigen-specific stimulation and significantly higher basal pSTAT5 in CLL patients treated with immunochemotherapy. Thus, we speculate that through the use of this pharmacodynamic tool, the efficacy of immunosuppressive drugs and their possible off-target effects can be assessed.
Collapse
Affiliation(s)
- Zlatko Roškar
- Department of Haematology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Mojca Dreisinger
- Department of Haematology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Primož Tič
- Department of Laboratory Diagnostics, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Evgenija Homšak
- Department of Laboratory Diagnostics, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Sebastjan Bevc
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Department of Nephrology, University Medical Center Maribor, 2000 Maribor, Slovenia
| | - Aleš Goropevšek
- Department of Laboratory Diagnostics, University Medical Centre Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
41
|
Yüksel B, Hızlı Deniz AA, Şahin F, Sahin K, Türkel N. Cannabinoid compounds in combination with curcumin and piperine display an anti-tumorigenic effect against colon cancer cells. Front Pharmacol 2023; 14:1145666. [PMID: 37180710 PMCID: PMC10169831 DOI: 10.3389/fphar.2023.1145666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023] Open
Abstract
Currently, use of cannabinoids is limited to improve adverse effects of chemotherapy and their palliative administration during treatment is curiously concomitant with improved prognosis and regressed progression in patients with different tumor types. Although, non-psychoactive cannabidiol (CBD) and cannabigerol (CBG) display antineoplastic effects by repressing tumor growth and angiogenesis both in cell line and animal models, their use as chemotherapeutic agents is awaiting further investigation. Both clinical and epidemiological evidence supported by experimental findings suggest that micronutrients such as curcumin and piperine may present a safer strategy in preventing tumorigenesis and its recurrence. Recent studies demonstrated that piperine potentiates curcumin's inhibitory effect on tumor progression via enhancing its delivery and therapeutic activity. In this study, we investigated a plausible therapeutic synergism of a triple combination of CBD/CBG, curcumin, and piperine in the colon adenocarcinoma using HCT116 and HT29 cell lines. Potential synergistic effects of various combinations including these compounds were tested by measuring cancer cell proliferation and apoptosis. Our findings revealed that different genetic backgrounds of HCT116 and HT29 cell lines resulted in divergent responses to the combination treatments. Triple treatment showed synergism in terms of exhibiting anti-tumorigenic effects by activating the Hippo YAP signaling pathway in the HCT116 cell line.
Collapse
Affiliation(s)
- Büşra Yüksel
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Ayşen Aslı Hızlı Deniz
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Kazim Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig, Türkiye
| | - Nezaket Türkel
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| |
Collapse
|
42
|
Husseiny EM, S Abulkhair H, El-Dydamony NM, Anwer KE. Exploring the cytotoxic effect and CDK-9 inhibition potential of novel sulfaguanidine-based azopyrazolidine-3,5-diones and 3,5-diaminoazopyrazoles. Bioorg Chem 2023; 133:106397. [PMID: 36753965 DOI: 10.1016/j.bioorg.2023.106397] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/30/2022] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
Regarding the structural analysis of variable effective CDK-9 suppressors, we record the design and synthesis of two new sets of sulfaguanidine-based azopyrazolidine-3,5-diones and 3,5-diaminoazopyrazoles with expected anticancer and CDK-9 inhibiting activity. In the designed molecules, the pyrazole ring and sulphaguanidine fragment were linked together for the first time through diazo linkers as they are expected to enhance the anticancer activity and CDK degrading interaction. All derivatives have been estimated regarding their cytotoxic activity toward three tumor cells where CDK overexpression has been reported (HePG2, HCT-116, and MCF-7). Among these, four derivatives VII, VIII, X, and XIII exerted potent cytotoxicity against the chosen tumor cells presenting IC50 range equal to 2.86-25.89 µM. As well cytotoxicity on non-cancer cells and CDK-9 inhibition assay have been also assessed for these candidates to evaluate their selectivity indices and enzyme inhibition. The 3,5-diaminopyrazole-1-carboxamide derivative XIII showed a superior combined profile as cytotoxic with high selectivity toward cancer cells (HePG2: IC50 = 6.57 µM, SI = 13.31; HCT-116: IC50 = 9.54 µM, SI = 9.16; MCF-7: IC50 = 7.97 µM, SI = 10.97). Accordingly, it has been chosen to evaluate its probable mechanistic effect both in vitro (via enzyme assay, apoptosis induction, and cell cycle study) as well as in silico (through molecular docking). Overall, this work introduces the 3,5-diaminopyrazole-1-carboxamide derivative XIII as a potent CDK-9 inhibitor candidate (IC50 = 0.16 µM) that merits further investigations for the management of breast, colorectal, and hepatic malignancies.
Collapse
Affiliation(s)
- Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt.
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11884, Cairo, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, International Coastal Road, New Damietta 34518, Egypt.
| | - Nehad M El-Dydamony
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Kurls E Anwer
- Chemistry Department, Faculty of Science, Ain Shams University 11566, Abbassia, Cairo, Egypt.
| |
Collapse
|
43
|
Pershina AG, Nevskaya KV, Morozov KR, Litviakov NV. Methods for assessing the effect of microRNA on stemness genes. BULLETIN OF SIBERIAN MEDICINE 2023. [DOI: 10.20538/1682-0363-2022-4-170-182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
According to the latest concepts, for micrometastasis to develop into macrometastasis, differentiated cancer cells must revert to a dedifferentiated state. Activation of stemness genes plays a key role in this transition. Suppression of stemness gene expression using microRNAs can become the basis for the development of effective anti-metastatic drugs. This article provides an overview of the existing methods for assessing the effect of microRNAs on stemness genes and cancer cell dedifferentiation.
Collapse
Affiliation(s)
| | | | | | - N. V. Litviakov
- Siberian State Medical University;
Cancer Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| |
Collapse
|
44
|
Gonzalez-Pastor R, Carrera-Pacheco SE, Zúñiga-Miranda J, Rodríguez-Pólit C, Mayorga-Ramos A, Guamán LP, Barba-Ostria C. Current Landscape of Methods to Evaluate Antimicrobial Activity of Natural Extracts. Molecules 2023; 28:1068. [PMID: 36770734 PMCID: PMC9920787 DOI: 10.3390/molecules28031068] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/24/2023] Open
Abstract
Natural extracts have been and continue to be used to treat a wide range of medical conditions, from infectious diseases to cancer, based on their convenience and therapeutic potential. Natural products derived from microbes, plants, and animals offer a broad variety of molecules and chemical compounds. Natural products are not only one of the most important sources for innovative drug development for animal and human health, but they are also an inspiration for synthetic biology and chemistry scientists towards the discovery of new bioactive compounds and pharmaceuticals. This is particularly relevant in the current context, where antimicrobial resistance has risen as a global health problem. Thus, efforts are being directed toward studying natural compounds' chemical composition and bioactive potential to generate drugs with better efficacy and lower toxicity than existing molecules. Currently, a wide range of methodologies are used to analyze the in vitro activity of natural extracts to determine their suitability as antimicrobial agents. Despite traditional technologies being the most employed, technological advances have contributed to the implementation of methods able to circumvent issues related to analysis capacity, time, sensitivity, and reproducibility. This review produces an updated analysis of the conventional and current methods to evaluate the antimicrobial activity of natural compounds.
Collapse
Affiliation(s)
- Rebeca Gonzalez-Pastor
- Biomedical Research Center (CENBIO), Eugenio Espejo School of Health Sciences, Universidad UTE, Quito 170527, Ecuador
| | - Saskya E. Carrera-Pacheco
- Biomedical Research Center (CENBIO), Eugenio Espejo School of Health Sciences, Universidad UTE, Quito 170527, Ecuador
| | - Johana Zúñiga-Miranda
- Biomedical Research Center (CENBIO), Eugenio Espejo School of Health Sciences, Universidad UTE, Quito 170527, Ecuador
| | - Cristina Rodríguez-Pólit
- Biomedical Research Center (CENBIO), Eugenio Espejo School of Health Sciences, Universidad UTE, Quito 170527, Ecuador
| | - Arianna Mayorga-Ramos
- Biomedical Research Center (CENBIO), Eugenio Espejo School of Health Sciences, Universidad UTE, Quito 170527, Ecuador
| | - Linda P. Guamán
- Biomedical Research Center (CENBIO), Eugenio Espejo School of Health Sciences, Universidad UTE, Quito 170527, Ecuador
| | - Carlos Barba-Ostria
- School of Medicine, College of Health Sciences, Universidad San Francisco de Quito (USFQ), Quito 170901, Ecuador
| |
Collapse
|
45
|
Esimbekova AR, Palkina NV, Zinchenko IS, Belenyuk VD, Savchenko AA, Sergeeva EY, Ruksha T. Focal adhesion alterations in
G0
‐positive melanoma cells. Cancer Med 2022; 12:7294-7308. [PMID: 36533319 PMCID: PMC10067123 DOI: 10.1002/cam4.5510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Melanoma is a highly heterogeneous malignant tumor that exhibits various forms of drug resistance. Recently, reversal transition of cancer cells to the G0 phase of the cell cycle under the influence of therapeutic drugs has been identified as an event associated with tumor dissemination. In the present study, we investigated the ability of chemotherapeutic agent dacarbazine to induce a transition of melanoma cells to the G0 phase as a mechanism of chemoresistance. METHODS We used the flow cytometry to analyze cell distribution within cell cycle phases after dacarbazine treatment as well as to identifyG0 -positive cells population. Transcriptome profiling was provided to determine genes associated with dacarbazine resistance. We evaluated the activity of β-galactosidase in cells treated with dacarbazine by substrate hydrolysis. Cell adhesion strength was measured by centrifugal assay application with subsequent staining of adhesive cells with Ki-67 monoclonal antibodies. Ability of melanoma cells to metabolize dacarbazine was determined by expressional analysis of CYP1A1, CYP1A2, CYP2E1 followed by CYP1A1 protein level evaluation by the ELISA method. RESULTS The present study determined that dacarbazine treatment of melanoma cells could induce an increase in the percentage of cells in G0 phase without alterations of β-galactosidase positive cells which corresponded to the fraction of the senescent cells. Transcriptomic profiling of cells under dacarbazine induction of G0 -positive cells percentage revealed that 'VEGFA-VEGFR2 signaling pathway' and 'Cell cycle' signaling were mostly enriched by dysregulated genes. 'Focal adhesion' signaling was also found to be triggered by dacarbazine. In melanoma cells treated with dacarbazine, an increase in G0 -positive cells among adherent cells was found. CONCLUSIONS Dacarbazine induces the alteration in a percentage of melanoma cells residing in G0 phase of a cell cycle. The altered adhesive phenotype of cancer cells under transition in the G0 phase may refer to a specific intercellular communication pattern of quiescent/senescent cancer cells.
Collapse
Affiliation(s)
| | - Nadezhda V. Palkina
- Department of Pathophysiology Krasnoyarsk State Medical University Krasnoyarsk Russia
| | - Ivan S. Zinchenko
- Department of Pathophysiology Krasnoyarsk State Medical University Krasnoyarsk Russia
| | - Vasiliy D. Belenyuk
- Laboratory of Cell Molecular Physiology and Pathology Federal Research Center, Krasnoyarsk Science Center of The Siberian Branch of The Russian Academy of Sciences Krasnoyarsk Russia
| | - Andrey A. Savchenko
- Laboratory of Cell Molecular Physiology and Pathology Federal Research Center, Krasnoyarsk Science Center of The Siberian Branch of The Russian Academy of Sciences Krasnoyarsk Russia
| | - Ekaterina Yu Sergeeva
- Department of Pathophysiology Krasnoyarsk State Medical University Krasnoyarsk Russia
| | - Tatiana G. Ruksha
- Department of Pathophysiology Krasnoyarsk State Medical University Krasnoyarsk Russia
| |
Collapse
|
46
|
Mishra HK. Clinical Applications of Flow Cytometry in Cancer Immunotherapies: From Diagnosis to Treatments. Methods Mol Biol 2022; 2593:93-112. [PMID: 36513926 DOI: 10.1007/978-1-0716-2811-9_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The scope of flow cytometry is rapidly expanding in the diagnosis of various cancers, and it is being used routinely as an aid in classifying leukemias and lymphomas. There are several applications of flow cytometry to enumerate tumorigenic anomalies in patients. The unusual distribution of cells in various locations, their DNA content, cell proliferation rate, dysregulated expression of several surface receptors, and expression of tumor antigens are some examples that can be characterized by using different flow cytometry-based techniques. For instance, the differential diagnosis between chronic lymphocytic leukemia (CLL) and various other mature B-cell neoplasms can be made by immunophenotyping in combination with absolute counting of numerous cellular subsets or by enumerating their percent distributions. Flow cytometry has several advantages over conventional techniques which include the ability to acquire a multiparametric data in a relatively shorter time and facilitate the comparative analysis of specific cellular subsets in an efficient manner.In addition to diagnosis, there are several other applications of flow cytometry in the management of various cancers which include treatment monitoring or even selecting a personalized precision-based immunotherapy in synch with advanced genetic tests to increase the chances of favorable prognosis and complete remission. The detection of chimeric antigen receptors (CARs) on various engineered effector cells can also be determined along with their specificity in engaging the targets. Furthermore, the assessment of numerous immunological parameters, their effector functions and potencies including the proliferation dynamics, cytokine secretion profiles, and activation efficiencies can also be measured before starting immunotherapies in patients.This chapter is a brief overview of flow cytometry applications in the diagnosis and treatment strategies of various cancers.
Collapse
|
47
|
Tawfik HO, Belal A, Abourehab MAS, Angeli A, Bonardi A, Supuran CT, El-Hamamsy MH. Dependence on linkers' flexibility designed for benzenesulfonamides targeting discovery of novel hCA IX inhibitors as potent anticancer agents. J Enzyme Inhib Med Chem 2022; 37:2765-2785. [PMID: 36210545 PMCID: PMC9559471 DOI: 10.1080/14756366.2022.2130285] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Herein we reported the design and synthesis of two series comprising twenty-two benzenesulfonamides that integrate the s-triazine moiety. Target compounds successfully suppressed the hCA IX, with IC50 ranging from 28.6 to 871 nM. Compounds 5d, 11b, 5b, and 7b were the most active analogues, which inhibited hCA IX isoform in the low nanomolar range (KI = 28.6, 31.9, 33.4, and 36.6 nM, respectively). Furthermore, they were assessed for their cytotoxic activity against a panel of 60 cancer cell lines following US-NCI protocol. According to five-dose assay, 13c showed significant anticancer activity than 5c with GI50-MID values of 25.08 and 189.01 µM, respectively. Additionally, 13c's effects on wound healing, cell cycle disruption, and apoptosis induction in NCI-H460 cancer cells were examined. Further, docking studies combined with molecular dynamic simulation showed a stable complex with high binding affinity of 5d to hCA IX, exploiting a favourable H-bond and lipophilic interactions.HIGHLIGHTSCarbonic anhydrase (CA) inhibitors comprising rigid and flexible linkers were developed.Compound 5d is the most potent CA IX inhibitor in the study (IC50: 28.6 nM).Compounds 5c and 13c displayed the greatest antiproliferative activity towards 60 cell lines.Compound 13c exposed constructive outcomes on normal cell lines, metastasis, and wound healing.Molecular docking and molecular dynamics (MDs) simulation was utilised to study binding mode.
Collapse
Affiliation(s)
- Haytham O. Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt,CONTACT H. O. Tawfik Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amany Belal
- Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt,Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Mohammed A. S. Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia,Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia, Egypt
| | - Andrea Angeli
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Alessandro Bonardi
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Claudiu T. Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy,C. T. Supuran Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Mervat H. El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
48
|
Woranush W, Moskopp ML, Noll T, Dieterich P. Quantifying and mathematical modelling of the influence of soluble adenylate cyclase on cell cycle in human endothelial cells with Bayesian inference. J Cell Mol Med 2022; 26:5887-5900. [DOI: 10.1111/jcmm.17611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 08/28/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Warunya Woranush
- Institut für Physiologie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden Dresden Germany
| | - Mats Leif Moskopp
- Institut für Physiologie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden Dresden Germany
- Vivantes Klinikum im Friedrichshain, Charité Academic Teaching Hospital, Klinik für Neurochirurgie Berlin Germany
| | - Thomas Noll
- Institut für Physiologie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden Dresden Germany
| | - Peter Dieterich
- Institut für Physiologie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden Dresden Germany
| |
Collapse
|
49
|
Bora D, Sharma A, John SE, Shankaraiah N. Development of hydrazide hydrazone-tethered combretastatin-oxindole derivatives as antimitotic agents. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
Yamada D, Vu S, Wu X, Shi Z, Morris D, Bloomstein JD, Huynh M, Zheng J, Hwang ST. Gain-of-function of TRPM4 predisposes mice to psoriasiform dermatitis. Front Immunol 2022; 13:1025499. [PMID: 36341417 PMCID: PMC9632438 DOI: 10.3389/fimmu.2022.1025499] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022] Open
Abstract
Transient receptor potential melastatin 4 (TRPM4) is a Ca2+-activated, monovalent cation channel that is expressed in a wide range of cells. We previously reported two gain-of-function (GoF) mutations of TRPM4 as the cause of progressive symmetric erythrokeratodermia (PSEK), which shares similar clinical and histopathological features with psoriasis. Using CRISPR/Cas9 technology, we generated TRPM4I1029M mice that have the equivalent mutation to one of the two genetic mutations found in human PSEK (equivalent to human TRPM4I1033M). Using this mutant mice, we examined the effects of TRPM4 GoF at the cellular and phenotypic levels to elucidate the pathological mechanisms underlying PSEK. In the absence of experimental stimulation, TRPM4I1029M mice did not show a phenotype. When treated with imiquimod (IMQ), however, TRPM4I1029M mice were predisposed to more severe psoriasiform dermatitis (PsD) than wild-type (WT), which was characterized by greater accumulation of CCR6-expressing γδ T cells and higher mRNA levels of Il17a. In TRPM4I1029M mice, dendritic cells showed enhanced migration and keratinocytes exhibited increased proliferation. Moreover, a TRPM4 inhibitor, glibenclamide, ameliorated PsD in WT and TRPM4I1029M mice. Our results indicate elevated TRPM4 activities boosted susceptibility to cutaneous stimuli, likely through elevation of membrane potential and alteration of downstream cellular signaling, resulting in enhanced inflammation. Our results further suggest a possible therapeutic application of TRPM4 inhibitors in psoriasis.
Collapse
Affiliation(s)
- Daisuke Yamada
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| | - Simon Vu
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Xuesong Wu
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| | - Zhenrui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Desiree Morris
- Kirk Kerkorian School of Medicine at University of Nevada, Las Vegas, Las, Vegas, NV, United States
| | - Joshua D Bloomstein
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| | - Mindy Huynh
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| | - Jie Zheng
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Samuel T Hwang
- Department of Dermatology, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|