1
|
Lovisa S, Vetrano S. TWISTed fibroblasts: New drivers of intestinal fibrosis in Crohn's disease. Heliyon 2024; 10:e40604. [PMID: 39654763 PMCID: PMC11626011 DOI: 10.1016/j.heliyon.2024.e40604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
Fibrosis is the pathological consequence of chronic inflammation. In Crohn's disease (CD), fibrostenotic complications occur with 50-70 % frequency as a failure to properly repair the tissue damage. Intestinal stenosis requires surgical intervention and relapses in most patients. Mesenchymal cells encompassed of heterogeneous cell subsets orchestrate this complex process. The lack of a full characterization of the stromal diversity and function in CD has consequently slowed the development of anti-fibrotic targets. Two recent studies align together demonstrating FAP+TWIST1+ fibroblasts as the primary mesenchymal population driving intestinal fibrosis in CD. Genetic and pharmacological targeting of Twist1 in mouse models proved the functional role of Fap+Twist1+ fibroblasts and indicate the use of the Twist1 inhibitor harmine as a potential therapeutic strategy to revert fibrosis.
Collapse
Affiliation(s)
- Sara Lovisa
- Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
- Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Stefania Vetrano
- Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
- Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
2
|
Zamir Nasta T, Tabandeh MR, Abbasi A, Moradi H, Imani MM, Jalili C. Harmine promotes odontoblastic differentiation of dental pulp stem cells. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024:1-12. [PMID: 39540882 DOI: 10.1080/15257770.2024.2427930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 07/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Dental pulp stem cells (DPSCs) have the potential to differentiate into various types of tissues including tooth, adipose, cartilage, muscle, nerve, and also possess regenerative properties. Harmine, a beta-carboline alkaloid, has been shown to have antitumor activities and promote bone formation through the differentiation of osteoblasts. The aim of this study was to investigate the effect of harmine on the differentiation of DPSCs into odontoblast cells. MATERIALS AND METHODS DPSCs were obtained from Iran's National Genetic Reserve Center and cultured under standard stem cell culture conditions. The cells were differentiated in culture medium with and without harmine, and cell viability was evaluated using MTT assay at different harmine concentrations. Moreover, differentiation of cells was measured using Alizarin Red staining, and the expression of Runx2, DSPP, and DMP1 genes was evaluated using western blotting and real-time PCR. RESULTS Harmine increased the survival rate of DPSCs in a time--dependent manner, but higher doses (above 80 μM) had a toxic effect. On day 14, Alizarin Red staining showed increased differentiation of odontoblasts in the harmine-treated groups compared to the untreated groups. Furthermore, harmine increased the expression of Runx2, DSPP, and DMP1 genes and proteins. CONCLUSION These findings suggest that harmine has a significant impact on the differentiation and proliferation of odontoblasts in DPSCs, likely due to its various properties and role in healing various diseases. Therefore, harmine could serve as a potential therapeutic agent for promoting dental tissue regeneration using DPSCs.
Collapse
Affiliation(s)
- Touraj Zamir Nasta
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Tabandeh
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Stem Cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hiva Moradi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Moslem Imani
- Department of Orthodontic, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Cyrus Jalili
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
3
|
Wang Y, Han D, Chai L, Qiu Y, Liu J, Li D, Zhang Q, Shen N, Chen Y, Chen H, Zhang J, Wang Q, Wang J, Li S, Xie X, Li M. MFN2-dependent mitochondrial dysfunction contributes to Relm-β-induced pulmonary arterial hypertension via USP18/Twist1/miR-214 pathway. Eur J Pharmacol 2024; 980:176828. [PMID: 39094924 DOI: 10.1016/j.ejphar.2024.176828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/03/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Induction of resistin-like molecule β (Relm-β) and mitofusin 2 (MFN2) mediated aberrant mitochondrial fission have been found to be involved in the pathogenesis of pulmonary arterial hypertension (PAH). However, the molecular mechanisms underlying Relm-β regulation of MFN2 therefore mitochondrial fission remain unclear. This study aims to address these issues. Primary cultured PASMCs and monocrotaline (MCT)-induced PAH rats were applied in this study. The results showed that Relm-β promoted cells proliferation in PASMCs, this was accompanied with the upregulation of USP18, Twist1 and miR-214, and downregulation of MFN2. We found that Relm-β increased USP18 expression which in turn raised Twist1 by suppressing its proteasome degradation. Elevation of Twist1 increased miR-214 expression and then reduced MFN2 expression and mitochondrial fragmentation leading to PASMCs proliferation. In vivo study, we confirmed that Relm-β was elevated in MCT-induced PAH rat model, and USP18/Twist1/miR-214/MFN2 axis was altered similar as in vitro. Targeting this cascade by Relm-β receptor inhibitor Calhex231, proteasome inhibitor MG-132, Twist1 inhibitor Harmine or miR-214 antagomiR prevented the development of pulmonary vascular remodeling and therefore PAH in MCT-treated rats. In conclusion, we demonstrate that Relm-β promotes PASMCs proliferation and vascular remodeling by activating USP18/Twist1/miR-214 dependent MFN2 reduction and mitochondrial fission, suggesting that this signaling pathway might be a promising target for management of PAH.
Collapse
Affiliation(s)
- Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Dong Han
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, PR China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yuanjie Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Nirui Shen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yuqian Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Huan Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jia Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
4
|
Wang Y, Chen SJ, Ma T, Long Q, Chen L, Xu KX, Cao Y. Promotion of apoptosis in melanoma cells by taxifolin through the PI3K/AKT signaling pathway: Screening of natural products using WGCNA and CMAP platforms. Int Immunopharmacol 2024; 138:112517. [PMID: 38924866 DOI: 10.1016/j.intimp.2024.112517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/26/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024]
Abstract
Melanoma is a skin cancer originating from melanocytes. The global incidence rate of melanoma is rapidly increasing, posing significant public health challenges. Identifying effective therapeutic agents is crucial in addressing this growing problem. Natural products have demonstrated promising anti-tumor activity. In this study, a plant flavonoid, taxifolin, was screened using Weighted Correlation Network Analysis (WGCNA) in combination with the Connectivity Map (CMAP) platform. Taxifolin was confirmed to inhibit the proliferation, migration, and invasion ability of melanoma A375 and MV-3 cells by promoting apoptosis. Additionally, it suppressed the Epithelial-Mesenchymal Transition (EMT) process of melanoma cells. Cyber pharmacological analysis revealed that taxifolin exerts its inhibitory effect on melanoma through the PI3K/AKT signaling pathway, specifically by downregulating the protein expression of p-PI3K and p-AKT. Notably, the addition of SC-79, an activator of the PI3K/AKT signaling pathway, reversed the effects of taxifolin on cell migration and apoptosis. Furthermore, in vivo experiments demonstrated that taxifolin treatment slowed tumor growth in mice without significant toxic effects. Based on these findings, taxifolin holds promise as a potential drug for melanoma treatment.
Collapse
Affiliation(s)
- Ye Wang
- School of Clinical Medicine, Guizhou Medical University, No.9 Beijing Road, Yunyan District, Guiyang 550004, Guizhou, China
| | - Shao-Jie Chen
- School of Clinical Medicine, Guizhou Medical University, No.9 Beijing Road, Yunyan District, Guiyang 550004, Guizhou, China; Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, No.28 Gui Medical Street, Yunyan District, Guiyang 550004, Guizhou, China
| | - Ting Ma
- School of Clinical Medicine, Guizhou Medical University, No.9 Beijing Road, Yunyan District, Guiyang 550004, Guizhou, China
| | - Qiu Long
- School of Clinical Medicine, Guizhou Medical University, No.9 Beijing Road, Yunyan District, Guiyang 550004, Guizhou, China
| | - Lan Chen
- School of Clinical Medicine, Guizhou Medical University, No.9 Beijing Road, Yunyan District, Guiyang 550004, Guizhou, China
| | - Ke-Xin Xu
- School of Clinical Medicine, Guizhou Medical University, No.9 Beijing Road, Yunyan District, Guiyang 550004, Guizhou, China
| | - Yu Cao
- School of Clinical Medicine, Guizhou Medical University, No.9 Beijing Road, Yunyan District, Guiyang 550004, Guizhou, China; Department of Dermatology, Affiliated Hospital of Guizhou Medical University, No.28 Gui Medical Street, Yunyan District, Guiyang 550004, Guizhou, China.
| |
Collapse
|
5
|
Ke BJ, Abdurahiman S, Biscu F, Zanella G, Dragoni G, Santhosh S, De Simone V, Zouzaf A, van Baarle L, Stakenborg M, Bosáková V, Van Rymenant Y, Verhulst E, Verstockt S, Klein E, Bislenghi G, Wolthuis A, Frič J, Breynaert C, D’Hoore A, Van der Veken P, De Meester I, Lovisa S, Hawinkels LJ, Verstockt B, De Hertogh G, Vermeire S, Matteoli G. Intercellular interaction between FAP+ fibroblasts and CD150+ inflammatory monocytes mediates fibrostenosis in Crohn's disease. J Clin Invest 2024; 134:e173835. [PMID: 39042469 PMCID: PMC11324301 DOI: 10.1172/jci173835] [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: 07/24/2023] [Accepted: 06/14/2024] [Indexed: 07/25/2024] Open
Abstract
Crohn's disease (CD) is marked by recurring intestinal inflammation and tissue injury, often resulting in fibrostenosis and bowel obstruction, necessitating surgical intervention with high recurrence rates. To elucidate the mechanisms underlying fibrostenosis in CD, we analyzed the transcriptome of cells isolated from the transmural ileum of patients with CD, including a trio of lesions from each patient: non-affected, inflamed, and stenotic ileum samples, and compared them with samples from patients without CD. Our computational analysis revealed that profibrotic signals from a subset of monocyte-derived cells expressing CD150 induced a disease-specific fibroblast population, resulting in chronic inflammation and tissue fibrosis. The transcription factor TWIST1 was identified as a key modulator of fibroblast activation and extracellular matrix (ECM) deposition. Genetic and pharmacological inhibition of TWIST1 prevents fibroblast activation, reducing ECM production and collagen deposition. Our findings suggest that the myeloid-stromal axis may offer a promising therapeutic target to prevent fibrostenosis in CD.
Collapse
Affiliation(s)
- Bo-Jun Ke
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Saeed Abdurahiman
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Francesca Biscu
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Gaia Zanella
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Gabriele Dragoni
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Gastroenterology Research Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Sneha Santhosh
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Veronica De Simone
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Anissa Zouzaf
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Lies van Baarle
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Michelle Stakenborg
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Veronika Bosáková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| | - Yentl Van Rymenant
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Emile Verhulst
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sare Verstockt
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Elliott Klein
- Department of Immunology and Respiratory Research, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Gabriele Bislenghi
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Albert Wolthuis
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jan Frič
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Christine Breynaert
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Andre D’Hoore
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | | | - Ingrid De Meester
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sara Lovisa
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Lukas J.A.C. Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, Netherlands
| | - Bram Verstockt
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Gert De Hertogh
- Laboratory of Pathology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Gianluca Matteoli
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Zhang Y, Wang J, Sun H, Xun Z, He Z, Zhao Y, Qi J, Sun S, Yang Q, Gu Y, Zhang L, Zhou C, Ye Y, Wu N, Zou D, Su B. TWIST1+FAP+ fibroblasts in the pathogenesis of intestinal fibrosis in Crohn's disease. J Clin Invest 2024; 134:e179472. [PMID: 39024569 PMCID: PMC11405050 DOI: 10.1172/jci179472] [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: 01/24/2024] [Accepted: 07/17/2024] [Indexed: 07/20/2024] Open
Abstract
Intestinal fibrosis, a severe complication of Crohn's disease (CD), is characterized by excessive extracellular matrix (ECM) deposition and induces intestinal strictures, but there are no effective antifibrosis drugs available for clinical application. We performed single-cell RNA sequencing (scRNA-Seq) of fibrotic and nonfibrotic ileal tissues from patients with CD with intestinal obstruction. Analysis revealed mesenchymal stromal cells (MSCs) as the major producers of ECM and the increased infiltration of its subset FAP+ fibroblasts in fibrotic sites, which was confirmed by immunofluorescence and flow cytometry. Single-cell transcriptomic profiling of chronic dextran sulfate sodium salt murine colitis model revealed that CD81+Pi16- fibroblasts exhibited transcriptomic and functional similarities to human FAP+ fibroblasts. Consistently, FAP+ fibroblasts were identified as the key subtype with the highest level of ECM production in fibrotic intestines. Furthermore, specific knockout or pharmacological inhibition of TWIST1, which was highly expressed by FAP+ fibroblasts, could significantly ameliorate fibrosis in mice. In addition, TWIST1 expression was induced by CXCL9+ macrophages enriched in fibrotic tissues via IL-1β and TGF-β signal. These findings suggest the inhibition of TWIST1 as a promising strategy for CD fibrosis treatment.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
| | - Jiaxin Wang
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
| | - Hongxiang Sun
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Ministry of Education Key Laboratory of Cell Death and Differentiation, and
| | - Zhenzhen Xun
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Ministry of Education Key Laboratory of Cell Death and Differentiation, and
| | - Zirui He
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yizhou Zhao
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
| | - Jingjing Qi
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Ministry of Education Key Laboratory of Cell Death and Differentiation, and
| | - Sishen Sun
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
| | - Qidi Yang
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
| | - Yubei Gu
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
| | - Ling Zhang
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
| | - Chunhua Zhou
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
| | - Youqiong Ye
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Ministry of Education Key Laboratory of Cell Death and Differentiation, and
| | - Ningbo Wu
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Ministry of Education Key Laboratory of Cell Death and Differentiation, and
| | - Duowu Zou
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
| | - Bing Su
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Ministry of Education Key Laboratory of Cell Death and Differentiation, and
| |
Collapse
|
7
|
Liu FH, Lin XC, Liu YW, Zhang TT, Zhang YB, Xie ZL, Zhan Y, Hu P. Harmine inhibits the proliferation and migration and promotes the apoptosis of colon cancer cells via inhibition of the FAK/AKT and ERK 1/2/CREB signaling pathways. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-13. [PMID: 39001813 DOI: 10.1080/10286020.2024.2361767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/23/2024] [Indexed: 07/15/2024]
Abstract
Harmine is present in a variety of medicinal plants, and its effects on colon cancer cells remain unclear. Here, we found that harmine exhibited significant inhibitory effects on the proliferation of colon cancer cells by inhibiting the phosphorylation levels of the FAK/AKT and ERK1/2/CREB. Furthermore, harmine also inhibited the migration of colon cancer cells and suppressed the expression levels of MMP-2, MMP-9, and VEGF. Additionally, harmine-induced apoptosis in colon cancer cells by regulating the expression of Bcl-2 and Bax. In conclusion, our findings suggest that harmine exerts a significant inhibitory effect on the development of colon cancer cells.
Collapse
Affiliation(s)
- Fu-Hong Liu
- Institute of Translational Medicine, Nanchang University, Nanchang 330001, China
| | - Xing-Cheng Lin
- Institute of Translational Medicine, Nanchang University, Nanchang 330001, China
| | - Yu-Wei Liu
- Institute of Translational Medicine, Nanchang University, Nanchang 330001, China
| | - Tian-Tian Zhang
- Institute of Translational Medicine, Nanchang University, Nanchang 330001, China
| | - Yang-Bo Zhang
- Department of Neurology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhuo-Long Xie
- Joint Program of Nanchang University and Queen Mary University of London, Nanchang 330001, China
| | - Yuan Zhan
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ping Hu
- Institute of Translational Medicine, Nanchang University, Nanchang 330001, China
| |
Collapse
|
8
|
Hu Y, Yu X, Yang L, Xue G, Wei Q, Han Z, Chen H. Research progress on the antitumor effects of harmine. Front Oncol 2024; 14:1382142. [PMID: 38590646 PMCID: PMC10999596 DOI: 10.3389/fonc.2024.1382142] [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: 02/05/2024] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Harmine is a naturally occurring β-carboline alkaloid originally isolated from Peganum harmala. As a major active component, harmine exhibits a broad spectrum of pharmacological properties, particularly remarkable antitumor effects. Recent mechanistic studies have shown that harmine can inhibit cancer cell proliferation and metastasis through epithelial-to-mesenchymal transition, cell cycle regulation, angiogenesis, and the induction of tumor cell apoptosis. Furthermore, harmine reduces drug resistance when used in combination with chemotherapeutic drugs. Despite its remarkable antitumor activity, the application of harmine is limited by its poor solubility and toxic side effects, particularly neurotoxicity. Novel harmine derivatives have demonstrated strong clinical application prospects, but further validation based on drug activity, acute toxicity, and other aspects is necessary. Here, we present a review of recent research on the action mechanism of harmine in cancer treatment and the development of its derivatives, providing new insights into its potential clinical applications and strategies for mitigating its toxicity while enhancing its efficacy.
Collapse
Affiliation(s)
- Yonghua Hu
- Key Laboratory of the Digestive System Tumors of Gansu Province, Department of Tumor Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xiaoli Yu
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Lei Yang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Gaimei Xue
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qinglin Wei
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhijian Han
- Key Laboratory of the Digestive System Tumors of Gansu Province, Department of Tumor Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Hao Chen
- Key Laboratory of the Digestive System Tumors of Gansu Province, Department of Tumor Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
9
|
Fakhri S, Moradi SZ, Faraji F, Kooshki L, Webber K, Bishayee A. Modulation of hypoxia-inducible factor-1 signaling pathways in cancer angiogenesis, invasion, and metastasis by natural compounds: a comprehensive and critical review. Cancer Metastasis Rev 2024; 43:501-574. [PMID: 37792223 DOI: 10.1007/s10555-023-10136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023]
Abstract
Tumor cells employ multiple signaling mediators to escape the hypoxic condition and trigger angiogenesis and metastasis. As a critical orchestrate of tumorigenic conditions, hypoxia-inducible factor-1 (HIF-1) is responsible for stimulating several target genes and dysregulated pathways in tumor invasion and migration. Therefore, targeting HIF-1 pathway and cross-talked mediators seems to be a novel strategy in cancer prevention and treatment. In recent decades, tremendous efforts have been made to develop multi-targeted therapies to modulate several dysregulated pathways in cancer angiogenesis, invasion, and metastasis. In this line, natural compounds have shown a bright future in combating angiogenic and metastatic conditions. Among the natural secondary metabolites, we have evaluated the critical potential of phenolic compounds, terpenes/terpenoids, alkaloids, sulfur compounds, marine- and microbe-derived agents in the attenuation of HIF-1, and interconnected pathways in fighting tumor-associated angiogenesis and invasion. This is the first comprehensive review on natural constituents as potential regulators of HIF-1 and interconnected pathways against cancer angiogenesis and metastasis. This review aims to reshape the previous strategies in cancer prevention and treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6714415153, Iran
| | - Kassidy Webber
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA.
| |
Collapse
|
10
|
Yang F, Akhtar MN, Zhang D, El-Mayta R, Shin J, Dorsey JF, Zhang L, Xu X, Guo W, Bagley SJ, Fuchs SY, Koumenis C, Lathia JD, Mitchell MJ, Gong Y, Fan Y. An immunosuppressive vascular niche drives macrophage polarization and immunotherapy resistance in glioblastoma. SCIENCE ADVANCES 2024; 10:eadj4678. [PMID: 38416830 PMCID: PMC10901371 DOI: 10.1126/sciadv.adj4678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Cancer immunity is subjected to spatiotemporal regulation by leukocyte interaction with neoplastic and stromal cells, contributing to immune evasion and immunotherapy resistance. Here, we identify a distinct mesenchymal-like population of endothelial cells (ECs) that form an immunosuppressive vascular niche in glioblastoma (GBM). We reveal a spatially restricted, Twist1/SATB1-mediated sequential transcriptional activation mechanism, through which tumor ECs produce osteopontin to promote immunosuppressive macrophage (Mφ) phenotypes. Genetic or pharmacological ablation of Twist1 reverses Mφ-mediated immunosuppression and enhances T cell infiltration and activation, leading to reduced GBM growth and extended mouse survival, and sensitizing tumor to chimeric antigen receptor T immunotherapy. Thus, these findings uncover a spatially restricted mechanism controlling tumor immunity and suggest that targeting endothelial Twist1 may offer attractive opportunities for optimizing cancer immunotherapy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Md Naushad Akhtar
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Junyoung Shin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jay F. Dorsey
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen J. Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin D. Lathia
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
11
|
Fakhri S, Moradi SZ, Abbaszadeh F, Faraji F, Amirian R, Sinha D, McMahon EG, Bishayee A. Targeting the key players of phenotypic plasticity in cancer cells by phytochemicals. Cancer Metastasis Rev 2024; 43:261-292. [PMID: 38169011 DOI: 10.1007/s10555-023-10161-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024]
Abstract
Plasticity of phenotypic traits refers to an organism's ability to change in response to environmental stimuli. As a result, the response may alter an organism's physiological state, morphology, behavior, and phenotype. Phenotypic plasticity in cancer cells describes the considerable ability of cancer cells to transform phenotypes through non-genetic molecular signaling activities that promote therapy evasion and tumor metastasis via amplifying cancer heterogeneity. As a result of metastable phenotypic state transitions, cancer cells can tolerate chemotherapy or develop transient adaptive resistance. Therefore, new findings have paved the road in identifying factors and agents that inhibit or suppress phenotypic plasticity. It has also investigated novel multitargeted agents that may promise new effective strategies in cancer treatment. Despite the efficiency of conventional chemotherapeutic agents, drug toxicity, development of resistance, and high-cost limit their use in cancer therapy. Recent research has shown that small molecules derived from natural sources are capable of suppressing cancer by focusing on the plasticity of phenotypic responses. This systematic, comprehensive, and critical review analyzes the current state of knowledge regarding the ability of phytocompounds to target phenotypic plasticity at both preclinical and clinical levels. Current challenges/pitfalls, limitations, and future perspectives are also discussed.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| | - Roshanak Amirian
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Dona Sinha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata, 700 026, West Bengal, India
| | - Emily G McMahon
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
12
|
Kato Y, Fukazawa T, Tanimoto K, Kanawa M, Kojima M, Saeki I, Kurihara S, Touge R, Hirohashi N, Okada S, Hiyama E. Achaete-scute family bHLH transcription factor 2 activation promotes hepatoblastoma progression. Cancer Sci 2024; 115:847-858. [PMID: 38183173 PMCID: PMC10921009 DOI: 10.1111/cas.16051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/16/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024] Open
Abstract
Achaete-scute family bHLH transcription factor 2 (ASCL2) is highly expressed in hepatoblastoma (HB) tissues, but its role remains unclear. Thus, biological changes in the HB cell line HepG2 in response to induced ASCL2 expression were assessed. ASCL2 expression was induced in HepG2 cells using the Tet-On 3G system, which includes doxycycline. Cell viability, proliferation activity, mobility, and stemness were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, colony-formation, migration, invasion, and sphere-formation assays. Quantitative reverse-transcription polymerase chain reaction was used to assess the expression of markers for proliferation (CCND1 and MYC), epithelial-mesenchymal transition (EMT; SNAI1, TWIST1, and ZEB1), mesenchymal-epithelial transition (CDH1), and stemness (KLF4, POU5F1, and SOX9). Compared with the non-induced HepG2 cells, cells with induced ASCL2 expression showed significant increases in viability, colony number, migration area (%), and sphere number on days 7, 14, 8, and 7, respectively, and invasion area (%) after 90 h. Furthermore, induction of ASCL2 expression significantly upregulated CCND1, MYC, POU5F1, SOX9, and KLF4 expression on days 2, 2, 3, 3, and 5, respectively, and increased the ratios of SNAI1, TWIST1, and ZEB1 to CDH1 on day 5. ASCL2 promoted the formation of malignant phenotypes in HepG2 cells, which may be correlated with the upregulation of the Wnt signaling pathway-, EMT-, and stemness-related genes. ASCL2 activation may therefore be involved in the progression of HB.
Collapse
Affiliation(s)
- Yutaka Kato
- Department of Pediatrics, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Takahiro Fukazawa
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
- Division of Medical Research Support, Advanced Research Support CenterEhime UniversityToonJapan
| | - Keiji Tanimoto
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and MedicineHiroshima UniversityHiroshimaJapan
| | - Masami Kanawa
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
| | - Masato Kojima
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| | - Isamu Saeki
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| | - Sho Kurihara
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| | - Ryo Touge
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| | - Nobuyuki Hirohashi
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and MedicineHiroshima UniversityHiroshimaJapan
| | - Satoshi Okada
- Department of Pediatrics, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Eiso Hiyama
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| |
Collapse
|
13
|
Bordeaux ZA, Reddy SV, Choi J, Braun G, McKeel J, Lu W, Yossef SM, Ma EZ, West CE, Kwatra SG, Kwatra MM. Transcriptomic and proteomic analysis of tumor suppressive effects of GZ17-6.02 against mycosis fungoides. Sci Rep 2024; 14:1955. [PMID: 38263212 PMCID: PMC10805783 DOI: 10.1038/s41598-024-52544-z] [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: 09/25/2023] [Accepted: 01/19/2024] [Indexed: 01/25/2024] Open
Abstract
Mycosis fungoides (MF) is the most common form of cutaneous T-cell lymphoma (CTCL). Despite having a wide variety of therapeutic agents available for the treatment of MF, patients often suffer from a significant decrease in quality of life and rarely achieve long-term remission or complete cure, highlighting a need to develop novel therapeutic agents for this disease. The present study was undertaken to evaluate the efficacy of a novel anti-tumor agent, GZ17-6.02, which is composed of curcumin, harmine, and isovanillin, against MF in vitro and in murine models. Treatment of HH and MyLa cells with GZ17-6.02 inhibited the growth of both cell lines with IC50 ± standard errors for growth inhibition of 14.37 ± 1.19 µg/mL and 14.56 ± 1.35 µg/mL, respectively, and increased the percentage of cells in late apoptosis (p = .0304 for HH; p = .0301 for MyLa). Transcriptomic and proteomic analyses revealed that GZ17-6.02 suppressed several pathways, including tumor necrosis factor (TNF)-ɑ signaling via nuclear factor (NF)-kB, mammalian target of rapamycin complex (mTORC)1, and Pi3K/Akt/mTOR signaling. In a subcutaneous tumor model, GZ17-6.02 decreased tumor volume (p = .002) and weight (p = .009) compared to control conditions. Proteomic analysis of tumor samples showed that GZ17-6.02 suppressed the expression of several proteins that may promote CTCL growth, including mitogen-activated protein kinase (MAPK)1, MAPK3, Growth factor receptor bound protein (GRB)2, and Mediator of RAP80 interactions and targeting subunit of 40 kDa (MERIT)40.
Collapse
Affiliation(s)
- Zachary A Bordeaux
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Sriya V Reddy
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Justin Choi
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Gabriella Braun
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Jaimie McKeel
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Weiying Lu
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Selina M Yossef
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Emily Z Ma
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
| | - Cameron E West
- Genzada Pharmaceuticals, Hutchinson, USA
- US Dermatology Partners, Wichita, USA
| | - Shawn G Kwatra
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - Madan M Kwatra
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, USA
| |
Collapse
|
14
|
Mirjat D, Kashif M, Roberts CM. Shake It Up Baby Now: The Changing Focus on TWIST1 and Epithelial to Mesenchymal Transition in Cancer and Other Diseases. Int J Mol Sci 2023; 24:17539. [PMID: 38139368 PMCID: PMC10743446 DOI: 10.3390/ijms242417539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
TWIST1 is a transcription factor that is necessary for healthy neural crest migration, mesoderm development, and gastrulation. It functions as a key regulator of epithelial-to-mesenchymal transition (EMT), a process by which cells lose their polarity and gain the ability to migrate. EMT is often reactivated in cancers, where it is strongly associated with tumor cell invasion and metastasis. Early work on TWIST1 in adult tissues focused on its transcriptional targets and how EMT gave rise to metastatic cells. In recent years, the roles of TWIST1 and other EMT factors in cancer have expanded greatly as our understanding of tumor progression has advanced. TWIST1 and related factors are frequently tied to cancer cell stemness and changes in therapeutic responses and thus are now being viewed as attractive therapeutic targets. In this review, we highlight non-metastatic roles for TWIST1 and related EMT factors in cancer and other disorders, discuss recent findings in the areas of therapeutic resistance and stemness in cancer, and comment on the potential to target EMT for therapy. Further research into EMT will inform novel treatment combinations and strategies for advanced cancers and other diseases.
Collapse
Affiliation(s)
- Dureali Mirjat
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Muhammad Kashif
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Cai M. Roberts
- Department of Pharmacology, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
15
|
Cordani N, Mologni L, Piazza R, Tettamanti P, Cogliati V, Mauri M, Villa M, Malighetti F, Di Bella C, Jaconi M, Cerrito MG, Cavaletti G, Lavitrano M, Cazzaniga ME. TWIST1 Upregulation Is a Potential Target for Reversing Resistance to the CDK4/6 Inhibitor in Metastatic Luminal Breast Cancer Cells. Int J Mol Sci 2023; 24:16294. [PMID: 38003483 PMCID: PMC10671583 DOI: 10.3390/ijms242216294] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Cyclin-dependent kinase (CDK) 4/6 inhibitors have significantly improved progression-free survival in hormone-receptor-positive (HR+), human-epidermal-growth-factor-receptor-type-2-negative (HER2-) metastatic luminal breast cancer (mLBC). Several studies have shown that in patients with endocrine-sensitive or endocrine-resistant LBC, the addition of CDK4/6 inhibitors to endocrine therapy significantly prolongs progression-free survival. However, the percentage of patients who are unresponsive or refractory to these therapies is as high as 40%, and no reliable and reproducible biomarkers have been validated to select a priori responders or refractory patients. The selection of mutant clones in the target oncoprotein is the main cause of resistance. Other mechanisms such as oncogene amplification/overexpression or mutations in other pathways have been described in several models. In this study, we focused on palbociclib, a selective CDK4/6 inhibitor. We generated a human MCF-7 luminal breast cancer cell line that was able to survive and proliferate at different concentrations of palbociclib and also showed cross-resistance to abemaciclib. The resistant cell line was characterized via RNA sequencing and was found to strongly activate the epithelial-to-mesenchymal transition. Among the top deregulated genes, we found a dramatic downregulation of the CDK4 inhibitor CDKN2B and an upregulation of the TWIST1 transcription factor. TWIST1 was further validated as a target for the reversal of palbociclib resistance. This study provides new relevant information about the mechanisms of resistance to CDK4/6 inhibitors and suggests potential new markers for patients' follow-up care during treatment.
Collapse
Affiliation(s)
- Nicoletta Cordani
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Luca Mologni
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Rocco Piazza
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Pietro Tettamanti
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Viola Cogliati
- Phase 1 Research Centre, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy (C.D.B.); (M.J.)
| | - Mario Mauri
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Matteo Villa
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Federica Malighetti
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Camillo Di Bella
- Phase 1 Research Centre, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy (C.D.B.); (M.J.)
| | - Marta Jaconi
- Phase 1 Research Centre, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy (C.D.B.); (M.J.)
| | - Maria Grazia Cerrito
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Guido Cavaletti
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
| | - Marina Elena Cazzaniga
- School of Medicine and Surgery, Milano-Bicocca University, 20900 Monza, Italy; (L.M.); (R.P.); (P.T.); (M.M.); (M.V.); (F.M.); (M.G.C.); (G.C.); (M.L.); (M.E.C.)
- Phase 1 Research Centre, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy (C.D.B.); (M.J.)
| |
Collapse
|
16
|
Kanoujia J, Das A, Raina N, Kaur G, Singh SK, Tuli HS, Garg A, Gupta M. Recent advances in BCRP-induced breast cancer resistance treatment with marine-based natural products. IUBMB Life 2023; 75:896-910. [PMID: 37439402 DOI: 10.1002/iub.2764] [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: 12/30/2022] [Accepted: 06/07/2023] [Indexed: 07/14/2023]
Abstract
Breast cancer is the prominent cause of cancer-related death in women globally in terms of incidence and mortality. Despite, recent advances in the management of breast cancer, there are still a lot of cases of resistance to medicines, which is currently one of the biggest problems faced by researchers across the globe. Out of several mechanisms, breast cancer resistance protein (BCRP) arbitrated drug resistance is a major concern. Hormonal, cytotoxic and immunotherapeutic drugs are used in the systemic therapy of breast cancer. It is vital to choose drugs based on the clinical and molecular attributes of the tumor to provide better treatment with greater efficacy and minimal harm. Given the aforementioned necessity, the use of marine flora in treating breast cancer cannot be neglected. The scientists also stressed the value of marine-derived goods in avoiding breast cancer resistance. Future research into the identification of anticancer drugs will heavily draw upon the marine environment's ample supply of marine-derived natural products (MNPs), which have a wide range of biological functions. Cell cycle arrest, induction of apoptosis and anti-angiogenic, anti-proliferative and anti-metastasis actions are all part of their processes. The overview of breast cancer, the mechanisms underlying its resistance, recent clinical trials based on marine-derived products in breast cancer and the use of marine products in the treatment of breast cancer are highlighted in this paper. Moreover, the authors also emphasised the importance of marine-derived products in preventing breast cancer resistance.
Collapse
Affiliation(s)
- Jovita Kanoujia
- Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, India
| | - Anjali Das
- Center of Ocean Research, Sathyabama Institute of Science and Technology, Chennai, India
| | - Neha Raina
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | | | - Hardeep S Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | - Ashish Garg
- Department of P.G. Studies and Research in Chemistry and Pharmacy, Rani Durgavati University Jabalpur, Jabalpur, India
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| |
Collapse
|
17
|
Fan R, Wang S, Wu Y, Feng Y, Gao M, Cao Y, Ma X, Xie S, Wang C, Gao L, Wang Y, Dai F. Activation of endoplasmic reticulum stress by harmine suppresses the growth of esophageal squamous cell carcinoma. Phytother Res 2023; 37:4655-4673. [PMID: 37525965 DOI: 10.1002/ptr.7933] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/16/2023] [Accepted: 06/21/2023] [Indexed: 08/02/2023]
Abstract
The worldwide overall 5-year survival rate of esophageal squamous cell carcinoma (ESCC) patients is less than 20%, and novel therapeutic strategies for these patients are urgently needed. Harmine is a natural β-carboline alkaloid, which received great interest in cancer research because of its biological and anti-tumor activities. The aim of this study is to examine the effects of harmine on ESCC and its mechanism. We investigated the effects of harmine on proliferation, cell cycle, apoptosis, and tumor growth in vivo. RNA sequencing (RNA-seq), real-time PCR, and western blotting were used to detect the mechanism. Harmine inhibited ESCC cell growth in vitro and tumor growth in vivo. Differentially expressed genes in harmine-treated ESCC cells were mainly involved in protein processing in the endoplasmic reticulum (ER). Real-time PCR and western blotting confirmed harmine-induced cellular ER stress. CRISPR-Cas9 knockout of C/EBP homologous protein (CHOP) abolished harmine-induced expression of death receptor 5 and apoptosis. Harmine also induced the expression of CHOP-mediated sestrin-2, which in turn contributes to autophagosome formation via suppressing the AMP-activated protein kinase-protein kinase B-mammalian target of rapamycin signaling pathway. In conclusion, our results demonstrate that harmine inhibits the growth of ESCC through its regulation of ER stress, suggesting that it is a promising candidate for ESCC treatment.
Collapse
Affiliation(s)
- Ronghui Fan
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Senzhen Wang
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yalan Wu
- School of Biomedical Sciences, Heart and Vascular Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yongli Feng
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Mengke Gao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Yue Cao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Xiaoxuan Ma
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Songqiang Xie
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Chaojie Wang
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Lei Gao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan University, Kaifeng, Henan, China
| | - Yanming Wang
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Fujun Dai
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| |
Collapse
|
18
|
Zani CP, Zani AP, Thomazini CM, Retamiro KM, de Oliveira AR, Gonçalves DL, Sarragiotto MH, Garcia FP, de Oliveira Silva S, Nakamura CV, Ueda-Nakamura T. β-Carboline-α-aminophosphonate Derivative: A Promising Antitumor Agent for Breast Cancer Treatment. Molecules 2023; 28:molecules28093949. [PMID: 37175359 PMCID: PMC10179861 DOI: 10.3390/molecules28093949] [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: 03/22/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Breast cancer is the most common type of cancer and the leading cause of cancer mortality among women worldwide. Considering the limitations of the current treatments available, we analyzed the in vitro cytotoxic potential of ((4-Fluoro-phenyl)-{2-[(1-phenyl-9H-β-carboline-3-carbonyl)-amino]-ethylamino}-methyl)-phosphonic acid dibutyl ester (BCP-1) in breast cancer cells (MCF-7 and MDA-MB-231) and in a non-tumor breast cell line (MCF-10A). BCP-1 has an α-aminophosphonate unit linked to the β-carboline nucleus, and the literature indicates that compounds of these classes have high biological potential. In the present study, the mechanism of action of BCP-1 was investigated through methods of spectrofluorimetry, flow cytometry, and protein expression analysis. It was found that BCP-1 inhibited the proliferation of both cancer cell lines. Furthermore, it induced oxidative stress and cell cycle arrest in G2/M. Upregulation of apoptosis-related proteins such as Bax, cytochrome C, and caspases, as well as a decrease in the anti-apoptotic protein Bcl-2, indicated potential induction of apoptosis in the MDA-MB-231 cells. While in MCF-7 cells, BCP-1 activated the autophagic death pathway, which was demonstrated by an increase in autophagic vacuoles and acidic organelles, in addition to increased expression of LC3I/LC3II and reduced SQSTM1/p62 expression. Further, BCP-1 demonstrated antimetastatic potential by reducing MMP-9 expression and cell migration in both breast cancer cell lines. In conclusion, BCP-1 is a promising candidate for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Caroline Pinto Zani
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Aline Pinto Zani
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Cristiane Melissa Thomazini
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Karina Miyuki Retamiro
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | | | - Débora Laís Gonçalves
- Department of Chemistry, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | | | - Francielle Pelegrin Garcia
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Sueli de Oliveira Silva
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Celso Vataru Nakamura
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Tania Ueda-Nakamura
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| |
Collapse
|
19
|
Tang X, Kurban M, Hafiz I, Shen Q, Wang M. Preparation of hyaluronic acid-loaded Harmine polymeric micelles and in vitro effect anti-breast cancer. Eur J Pharm Sci 2023; 183:106388. [PMID: 36758771 DOI: 10.1016/j.ejps.2023.106388] [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: 11/02/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 02/10/2023]
Abstract
AIMS To prepare hyaluronic acid-loaded Harmine polymeric micelles with CD44 targeting properties and to investigate their anti-breast cancer effects in vitro. METHODS The carboxyl group on hyaluronic acid is coupled to the amino group on 3,5-bis(trifluoromethyl)benzylamine by an amidation reaction. And the polymeric micelles self-assemble to encapsulate the Harmine in a hydrophobic core, characterized the polymer micelles by IR, 19F-NMR, Malvern particle sizing, release, hemolysis, and other experiments. Used CD44-positive MDA-MB-231 cells and CD44-negative MCF-7 cells as tumor models. The effect of polymer micelles on breast cancer cells in vitro by cytotoxicity assay, confocal, and flow cytometry. RESULTS The prepared polymer micelles had a uniform particle size of about 200 nm, good dispersion, PDI < 0.3, encapsulation rate up to 87%, drug loading of 4.12±0.03%, and negative charge. Hyaluronidase has a good enzymatic effect on polymeric micelles, with a hemolysis rate of less than 1%. It showed some dose-dependent toxicity to both MDA-MB-231 and MCF-7, with increased uptake of polymer micelles by CD44-positive MDA-MB-231 compared to CD44-negative MCF-7 cells and significant effects of polymer micelles on apoptosis and cycling in both cell types. These results suggest that the hyaluronic acid-loaded Harmine polymer micelles designed in this study are effective in killing breast cancer cells while at the same time reducing the toxicity of Harmine and improving its slow-release targeting.
Collapse
Affiliation(s)
- Xiaohui Tang
- Central Laboratory of Xinjiang Medical University, Urumqi 830017, China
| | - Munire Kurban
- Department of Pharmacy, Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, China
| | - Ipargul Hafiz
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Qi Shen
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Mei Wang
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
20
|
Hashemi M, Arani HZ, Orouei S, Fallah S, Ghorbani A, Khaledabadi M, Kakavand A, Tavakolpournegari A, Saebfar H, Heidari H, Salimimoghadam S, Entezari M, Taheriazam A, Hushmandi K. EMT mechanism in breast cancer metastasis and drug resistance: Revisiting molecular interactions and biological functions. Biomed Pharmacother 2022; 155:113774. [DOI: 10.1016/j.biopha.2022.113774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 12/24/2022] Open
|
21
|
Phytotherapeutic applications of alkaloids in treating breast cancer. Biomed Pharmacother 2022; 155:113760. [DOI: 10.1016/j.biopha.2022.113760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/12/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
|
22
|
Rashidi M, Mahmoudian E, Mirzaei S, Mazloomi SN, Bazi A, Azadeh H, Mozaffari M. Harmaline downregulates angiogenesis markers and suppresses the growth of 4T1 breast cancer cells in vivo and in vitro. Chem Biol Interact 2022; 365:110087. [PMID: 35963316 DOI: 10.1016/j.cbi.2022.110087] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 11/29/2022]
Abstract
The anti-angiogenic effects of harmaline, an alkaloid with emerging anti-tumor properties, are under investigation. In the present study, the effects of different doses of harmaline, either alone or in combination with doxorubicin (DOX), were assessed in mice models of breast tumor. Breast tumors were created by the subcutaneous injection of 4T1 cells into Balb/c mice. The mice received either normal saline, harmaline alone (10, 20, or 30 mg/kg), or harmaline (20 mg/kg) + DOX (10 mg/kg). Immunohistochemistry, ELISA, and real-time PCR were conducted to measure target parameters. Harmaline significantly increased tumor cells' sensitivity to DOX as confirmed by a significantly reduced tumor volume in the harmaline + DOX group after 24 days (P < 0.05). Also, the levels of Ki-67 (P < 0.001), MMP-2 (P < 0.001), and VEGF (P < 0.001) significantly decreased while the level of E-cadherin increased (P < 0.001) in the tumor tissues of the mice treated with 20 or 30 mg/kg harmaline or harmaline (20 mg/kg) + DOX (10 mg/kg) compared to the control group. There was a significant reduction in the serum level of IL-4 in tumor-bearing mice treated with harmaline (P < 0.05), and IFN-γ serum level was significantly augmented in all experimental groups compared to the control group (P < 0.05). The genes encoding VEGF, VEGF receptor 2, CD105, and COX2 were significantly down-regulated (P < 0.05 for all) in harmaline-treated (either alone or in combination with DOX) mice. In conclusion, harmaline seems to have the potential to be used as an anticancer agent for treating breast cancer.
Collapse
Affiliation(s)
- Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center,Mazandaran University of Medical Sciences, Sari, Iran.
| | - Elham Mahmoudian
- Cellular & Molecular Medicine Department, Faculty of Medicine, University of Ottawa, 451 ch. Smyth Rd., Roger Guindon Hall, Ottawa, ON, K1H 8M5, Canada
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Seyadeh Narges Mazloomi
- The Health of Plant and LivestockProducts Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Food and Drug Administration, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Bazi
- Department of Hematology, Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Hossein Azadeh
- Department of Internal Medicine, Rheumatology Division, Orthopedic Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mobina Mozaffari
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
23
|
Nasibova T. Cancer Statistics and Anticancer Potential of Peganum harmala Alkaloids: A Review. BORNEO JOURNAL OF PHARMACY 2022. [DOI: 10.33084/bjop.v5i1.3052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cancer is one of the most common diseases in the world. Although it develops in various organs and tissues, some species maintain a stable position in the ranking. Although the cancer causes are different, the specific grounds for each type are also noted. Sometimes the increase in incidents and mortality is associated with geographical reasons. Increases in statistics, expensive and chemotherapeutic methods focus on plant-based substances. One of such potential plants is Peganum harmala, which contains alkaloids such as harmine, harmaline, harmol, and harmalol. The effects of these compounds on many cancer cells have been tested, and positive results have been obtained. This fact reinforces the claim that more in-depth research on noted alkaloids is needed.
Collapse
|
24
|
Zhou R, Hu Z, Pan J, Wang J, Pei Y. Current research status of alkaloids against breast cancer. CHINESE J PHYSIOL 2022; 65:12-20. [DOI: 10.4103/cjp.cjp_89_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|