1
|
Dogan E, Galifi CA, Cecen B, Shukla R, Wood TL, Miri AK. Extracellular matrix regulation of cell spheroid invasion in a 3D bioprinted solid tumor-on-a-chip. Acta Biomater 2024; 186:156-166. [PMID: 39097123 PMCID: PMC11390304 DOI: 10.1016/j.actbio.2024.07.040] [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: 03/12/2024] [Revised: 07/01/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Tumor organoids and tumors-on-chips can be built by placing patient-derived cells within an engineered extracellular matrix (ECM) for personalized medicine. The engineered ECM influences the tumor response, and understanding the ECM-tumor relationship accelerates translating tumors-on-chips into drug discovery and development. In this work, we tuned the physical and structural characteristics of ECM in a 3D bioprinted soft-tissue sarcoma microtissue. We formed cell spheroids at a controlled size and encapsulated them into our gelatin methacryloyl (GelMA)-based bioink to make perfusable hydrogel-based microfluidic chips. We then demonstrated the scalability and customization flexibility of our hydrogel-based chip via engineering tools. A multiscale physical and structural data analysis suggested a relationship between cell invasion response and bioink characteristics. Tumor cell invasive behavior and focal adhesion properties were observed in response to varying polymer network densities of the GelMA-based bioink. Immunostaining assays and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) helped assess the bioactivity of the microtissue and measure the cell invasion. The RT-qPCR data showed higher expressions of HIF-1α, CD44, and MMP2 genes in a lower polymer density, highlighting the correlation between bioink structural porosity, ECM stiffness, and tumor spheroid response. This work is the first step in modeling STS tumor invasiveness in hydrogel-based microfluidic chips. STATEMENT OF SIGNIFICANCE: We optimized an engineering protocol for making tumor spheroids at a controlled size, embedding spheroids into a gelatin-based matrix, and constructing a perfusable microfluidic device. A higher tumor invasion was observed in a low-stiffness matrix than a high-stiffness matrix. The physical characterizations revealed how the stiffness is controlled by the density of polymer chain networks and porosity. The biological assays revealed how the structural properties of the gelatin matrix and hypoxia in tumor progression impact cell invasion. This work can contribute to personalized medicine by making more effective, tailored cancer models.
Collapse
Affiliation(s)
- Elvan Dogan
- Department of Biomedical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Christopher A Galifi
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Berivan Cecen
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Roshni Shukla
- Department of Biomedical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Amir K Miri
- Department of Biomedical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA; Department of Mechanical and Industrial Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| |
Collapse
|
2
|
He H, Dong K, Chen M, Wang Y, Li Y, Wang D, Jia M, Meng X, Sun W, Fu S, Yu J. TOB1 inhibits the gastric cancer progression by focal adhesion pathway and ERK pathway based on transcriptional and metabolic sequencing. BMC Cancer 2024; 24:1130. [PMID: 39261761 PMCID: PMC11389266 DOI: 10.1186/s12885-024-12894-3] [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: 04/01/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
Gastric cancer is one of the most malignant digestive tract tumors worldwide and its progression is associated with gene expression and metabolic alteration. We revealed that the gastric cancer patients with lower expression level of TOB1 exhibited poorer overall survivals according to the data in Kaplan-Meier Plotter. The unphosphorylated TOB1 protein which is effective expressed lower in gastric cancer cells. The gastric cancer cells with TOB1 gene depletion performed higher abilities of proliferation, migration and invasion and lower ability of apoptosis in vitro. The TOB1 gene depletion also promoted the tumorigenesis of gastric cancer cells in vivo. The gastric cancer cells with TOB1 gene overexpression had the converse behaviors. The transcriptional and metabolic sequencing was performed. The analyzation results showed that genes correlate-expressed with TOB1 gene were enriched in the pathways related to ERK pathway, including focal adhesion pathway, which was verified using real-time quantitative PCR. After inhibiting ERK pathway, the proliferation, colony formation and migration abilities were reduced in gastric cancer cells with low phosphorylated TOB1 protein expression level. Moreover, Pearson correlation analysis was adopted to further analyze the correlation of enriched metabolic products and differentially expressed genes. The expression of Choline, UDP-N-acetylglucosamine, Adenosine and GMP were related to the function of TOB1. This study demonstrates the genes and metabolites related to focal adhesion pathway and ERK pathway are the potential diagnosis and therapeutic targets to gastric cancer with TOB1 depletion.
Collapse
Affiliation(s)
- Hongjie He
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Kexian Dong
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, 150081, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Mingming Chen
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Yuanyuan Wang
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Yawen Li
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Dong Wang
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Mansha Jia
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Xiangning Meng
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, 150081, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Wenjing Sun
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, 150081, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Songbin Fu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, 150081, China
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Jingcui Yu
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China.
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
3
|
Li QY, Zhu RR, Yu HY, Liu CL, Diao FY, Jiang YQ, Lin YQ, Li XT, Wang WJ. Multifunctional targeting of docetaxel plus bakuchiol micelles in the treatment of invasion and metastasis of ovarian cancer. Biomed Mater 2024; 19:065002. [PMID: 39208838 DOI: 10.1088/1748-605x/ad7556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
The invasion and metastasis of tumors pose significant challenges in the treatment of ovarian cancer (OC), making it difficult to cure. One potential treatment approach that has gained attention is the use of matrix metalloproteinase reactive controlled release micelle preparations. In this study, we developed a novel PEG5000-PVGLIG-hyaluronic acid docetaxel/bakuchiol (PP-HA-DTX/BAK) micelles formulation with desirable characteristics such as particle size, narrow polydispersity index, and a ZETA potential of approximately -5 mV. The surface modification with HA facilitates tumor penetration into the tumor interior, while the incorporation of DSPE-PEG2000-PVGLIG-PEG5000helps conceal DSPE-PEG2000-HA, reducing off-target effects and prolonging drug circulation timein vivo. Bothin vitroandin vivoexperiments demonstrated that these micelles effectively inhibit proliferation, invasion, and metastasis of OC cells while promoting apoptosis. Therefore, our findings suggest that PP-HA-DTX/BAK micelles represent a safe and effective therapeutic strategy for treating OC.
Collapse
Affiliation(s)
- Qi-Yan Li
- Shandong Institute for Food and Drug Control, Shandong 250101, People's Republic of China
| | - Ri-Ran Zhu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong 250011, People's Republic of China
| | - Hai-Ying Yu
- Shandong Institute for Food and Drug Control, Shandong 250101, People's Republic of China
| | - Chun-Lin Liu
- Shandong Institute for Food and Drug Control, Shandong 250101, People's Republic of China
| | - Fei-Yan Diao
- Shandong Institute for Food and Drug Control, Shandong 250101, People's Republic of China
| | - Ya-Qi Jiang
- Shandong Institute for Food and Drug Control, Shandong 250101, People's Republic of China
| | - Yong-Qiang Lin
- Shandong Institute for Food and Drug Control, Shandong 250101, People's Republic of China
| | - Xue-Tao Li
- Liaoning University of Traditional Chinese Medicine, School of Pharmacy, Dalian 116600, People's Republic of China
| | - Wei-Jian Wang
- Shandong Institute for Food and Drug Control, Shandong 250101, People's Republic of China
| |
Collapse
|
4
|
Rozans S, Moghaddam AS, Wu Y, Atanasoff K, Nino L, Dunne K, Pashuck ET. Quantifying and Controlling the Proteolytic Degradation of Cell Adhesion Peptides. ACS Biomater Sci Eng 2024; 10:4916-4926. [PMID: 38968389 PMCID: PMC11322908 DOI: 10.1021/acsbiomaterials.4c00736] [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: 04/19/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024]
Abstract
Peptides are widely used within biomaterials to improve cell adhesion, incorporate bioactive ligands, and enable cell-mediated degradation of the matrix. While many of the peptides incorporated into biomaterials are intended to be present throughout the life of the material, their stability is not typically quantified during culture. In this work, we designed a series of peptide libraries containing four different N-terminal peptide functionalizations and three C-terminal functionalizations to better understand how simple modifications can be used to reduce the nonspecific degradation of peptides. We tested these libraries with three cell types commonly used in biomaterials research, including mesenchymal stem/stromal cells (hMSCs), endothelial cells, and macrophages, and quantified how these cell types nonspecifically degraded peptides as a function of terminal amino acid and chemistry. We found that peptides in solution which contained N-terminal amines were almost entirely degraded by 48 h, irrespective of the terminal amino acid, and that degradation occurred even at high peptide concentrations. Peptides with C-terminal carboxylic acids also had significant degradation when cultured with the cells. We found that simple modifications to the termini could significantly reduce or completely abolish nonspecific degradation when soluble peptides were added to cells cultured on tissue culture plastic or within hydrogel matrices, and that functionalizations which mimicked peptide conjugations to hydrogel matrices significantly slowed nonspecific degradation. We also found that there were minimal differences in peptide degradation across cell donors and that sequences mimicking different peptides commonly used to functionalize biomaterials all had significant nonspecific degradation. Finally, we saw that there was a positive trend between RGD stability and hMSC spreading within hydrogels, indicating that improving the stability of peptides within biomaterial matrices may improve the performance of engineered matrices.
Collapse
Affiliation(s)
- Samuel
J. Rozans
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Abolfazl Salehi Moghaddam
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Yingjie Wu
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Kayleigh Atanasoff
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Liliana Nino
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Katelyn Dunne
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - E. Thomas Pashuck
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| |
Collapse
|
5
|
Jagathesan K, Roy S. Recent Development of Transition Metal Complexes as Chemotherapeutic Hypoxia Activated Prodrug (HAP). ChemMedChem 2024; 19:e202400127. [PMID: 38634306 DOI: 10.1002/cmdc.202400127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024]
Abstract
Hypoxia is a state characterized by low concentration of Oxygen. Hypoxic state is often found in the central region of solid tumors. Hypoxia is associated with abnormal neovascularization resulted in poor blood flow in tissues and increased proliferation of tumor cells, imbalance between O2 supply and O2 consumption in tumor cells, high concentration of proton and strong reducibility. And, these abnormalities enhance the survival potency of the hypoxic tumours and increase the resistance towards chemotherapy and radiotherapy. One of the approach for treating hypoxic region of tumour is to use reducing environment of hypoxic tumours for reducing a molecule (hypoxia activated prodrug, HAP) and as a result the active drug will be released in hypoxic region in a controlled manner from the prodrug and kill the hypoxic tumour. Co(III) and Pt(IV) complexes with monodentate active drug molecule in the axial position can be reduced to Co(II) and Pt(II) moieties and as a result, the axial ligands (active drug) could come out from the metal center and could show its anticancer activity. In this review we have highlighted the research articles where transition metal-based complexes are used as chemotherapeutic hypoxia activated prodrug molecules which are reported in last 5 years.
Collapse
Affiliation(s)
- K Jagathesan
- Dept. of Chemistry, School of Advance Sciences, Vellore Institute of Technology, Vellore, 632014, India
| | - Sovan Roy
- Dept. of Chemistry, School of Advance Sciences, Vellore Institute of Technology, Vellore, 632014, India
| |
Collapse
|
6
|
Chang HF, Cheng JY. Glioblastoma U-87 cell electrotaxis is hindered by doxycycline with a concomitant reduction in the matrix metallopeptidase-9 expression. Biochem Biophys Rep 2024; 38:101690. [PMID: 38571555 PMCID: PMC10987802 DOI: 10.1016/j.bbrep.2024.101690] [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: 09/18/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
Electric fields (EF) play an essential role in cancer cell migration. Numerous cancer cell types exhibit electrotaxis under direct current electric fields (dcEF) of physiological electric field strength (EFs). This study investigated the effects of doxycycline on the electrotactic responses of U87 cells. After EF stimulation, U87 cells migrated toward the cathode, whereas doxycycline-treated U87 cells exhibited enhanced cell mobility but hindered cathodal migration. We further investigated the expression of the metastasis-correlated proteins matrix metallopeptidase-2 (MMP-2) and MMP-9 in U87 cells. The levels of MMP-2 in the cells were not altered under EF or doxycycline stimulation. In contrast, the EF stimulation greatly enhanced the levels of MMP-9 and then repressed in doxycycline-cotreated cells, accompanied by reduced cathodal migration. Our results demonstrated that an antibiotic at a non-toxic concentration could suppress the enhanced cell migration accelerated by EF of physiological strength. This finding may be applied as an anti-metastatic treatment for cancers.
Collapse
Affiliation(s)
- Hui-Fang Chang
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Ji-Yen Cheng
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Mechanical and Mechatronic Engineering, National Taiwan Ocean University, Keelung, Taiwan
- College of Engineering, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
7
|
Wehbe N, Badran A, Baydoun S, Al-Sawalmih A, Maresca M, Baydoun E, Mesmar JE. The Antioxidant Potential and Anticancer Activity of Halodule uninervis Ethanolic Extract against Triple-Negative Breast Cancer Cells. Antioxidants (Basel) 2024; 13:726. [PMID: 38929164 PMCID: PMC11200955 DOI: 10.3390/antiox13060726] [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: 04/28/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Natural remedies have been indispensable to traditional medicine practices for generations, offering therapeutic solutions for various ailments. In modern times, these natural products continue to play a pivotal role in the discovery of new drugs, especially for cancer treatment. The marine ecosystem offers a wide range of plants with potential anticancer activities due to their distinct biochemical diversity and adaptation to extreme situations. The seagrass Halodule uninervis is rich in diverse bioactive metabolites that bestow the plant with various pharmacological properties. However, its anticancer activity against invasive triple-negative breast cancer (TNBC) is still poorly investigated. In the present study, the phytochemical composition of an ethanolic extract of H. uninervis (HUE) was screened, and its antioxidant potential was evaluated. Moreover, the anticancer potential of HUE against MDA-MB-231 cells was investigated along with the possible underlying mechanisms of action. Our results showed that HUE is rich in diverse phytochemicals that are known for their antioxidant and anticancer effects. In MDA-MB-231 cells, HUE targeted the hallmarks of cancer, including cell proliferation, adhesion, migration, invasion, and angiogenesis. The HUE-mediated anti-proliferative and anti-metastatic effects were associated with the downregulation of the proto-oncogenic STAT3 signaling pathway. Taken together, H. uninervis could serve as a valuable source for developing novel drugs targeting TNBC.
Collapse
Affiliation(s)
- Nadine Wehbe
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107 2020, Lebanon; (N.W.); (E.B.)
| | - Adnan Badran
- Department of Nutrition, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 11196, Jordan;
| | - Serine Baydoun
- Breast Imaging Section, Imaging Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Ali Al-Sawalmih
- Marine Science Station, University of Jordan, Aqaba 11942, Jordan;
| | - Marc Maresca
- Aix-Marseille Univ, CNRS, Centrale Marseille, iSM2, 13013 Marseille, France
| | - Elias Baydoun
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107 2020, Lebanon; (N.W.); (E.B.)
| | - Joelle Edward Mesmar
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107 2020, Lebanon; (N.W.); (E.B.)
| |
Collapse
|
8
|
Bhuia MS, Chowdhury R, Akter MA, Ali MA, Afroz M, Akbor MS, Sonia FA, Mubarak MS, Islam MT. A mechanistic insight into the anticancer potentials of resveratrol: Current perspectives. Phytother Res 2024. [PMID: 38768953 DOI: 10.1002/ptr.8239] [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: 11/11/2023] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
Resveratrol is a widely recognized polyphenolic phytochemical found in various plants and their fruits, such as peanuts, grapes, and berry fruits. It is renowned for its several health advantages. The phytochemical is well known for its anticancer properties, and a substantial amount of clinical evidence has also established its promise as a chemotherapeutic agent. This study focuses on assessing the anticancer properties of resveratrol and gaining insight into the underlying molecular mechanisms. It also evaluates the biopharmaceutical, toxicological characteristics, and clinical utilization of resveratrol to determine its suitability for further development as a reliable anticancer agent. Therefore, the information about preclinical and clinical studies was collected from different electronic databases up-to-date (2018-2023). Findings from this study revealed that resveratrol has potent therapeutic benefits against various cancers involving different molecular mechanisms, such as induction of oxidative stress, cytotoxicity, inhibition of cell migration and invasion, autophagy, arresting of the S phase of the cell cycle, apoptotic, anti-angiogenic, and antiproliferative effects by regulating different molecular pathways including PI3K/AKT, p38/MAPK/ERK, NGFR-AMPK-mTOR, and so on. However, the compound has poor oral bioavailability due to reduced absorption; this limitation is overcome by applying nanotechnology (nanoformulation of resveratrol). Clinical application also showed therapeutic benefits in several types of cancer with no serious adverse effects. We suggest additional extensive studies to further check the efficacy, safety, and long-term hazards. This could involve a larger number of clinical samples to establish the compound as a reliable drug in the treatment of cancer.
Collapse
Affiliation(s)
- Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Dhaka, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Dhaka, Bangladesh
| | - Mst Asma Akter
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Md Arman Ali
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Meher Afroz
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Md Showkot Akbor
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Fatema Akter Sonia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Dhaka, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
9
|
Rozans SJ, Moghaddam AS, Wu Y, Atanasoff K, Nino L, Dunne K, Pashuck ET. Quantifying and controlling the proteolytic degradation of cell adhesion peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590329. [PMID: 38712239 PMCID: PMC11071418 DOI: 10.1101/2024.04.19.590329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Peptides are widely used within biomaterials to improve cell adhesion, incorporate bioactive ligands, and enable cell-mediated degradation of the matrix. While many of the peptides incorporated into biomaterials are intended to be present throughout the life of the material, their stability is not typically quantified during culture. In this work we designed a series of peptide libraries containing four different N-terminal peptide functionalizations and three C-terminal functionalization to better understand how simple modifications can be used to reduce non-specific degradation of peptides. We tested these libraries with three cell types commonly used in biomaterials research, including mesenchymal stem/stromal cells (hMSCs), endothelial cells, and macrophages, and quantified how these cell types non-specifically degraded peptide as a function of terminal amino acid and chemistry. We found that peptides in solution which contained N-terminal amines were almost entirely degraded by 48 hours, irrespective of the terminal amino acid, and that degradation occurred even at high peptide concentrations. Peptides with C-terminal carboxylic acids also had significant degradation when cultured with cells. We found that simple modifications to the termini could significantly reduce or completely abolish non-specific degradation when soluble peptides were added to cells cultured on tissue culture plastic or within hydrogel matrices, and that functionalizations which mimicked peptide conjugations to hydrogel matrices significantly slowed non-specific degradation. We also found that there were minimal differences across cell donors, and that sequences mimicking different peptides commonly-used to functionalized biomaterials all had significant non-specific degradation. Finally, we saw that there was a positive trend between RGD stability and hMSC spreading within hydrogels, indicating that improving the stability of peptides within biomaterial matrices may improve the performance of engineered matrices.
Collapse
|
10
|
Strippoli R, Niayesh-Mehr R, Adelipour M, Khosravi A, Cordani M, Zarrabi A, Allameh A. Contribution of Autophagy to Epithelial Mesenchymal Transition Induction during Cancer Progression. Cancers (Basel) 2024; 16:807. [PMID: 38398197 PMCID: PMC10886827 DOI: 10.3390/cancers16040807] [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: 12/15/2023] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Epithelial Mesenchymal Transition (EMT) is a dedifferentiation process implicated in many physio-pathological conditions including tumor transformation. EMT is regulated by several extracellular mediators and under certain conditions it can be reversible. Autophagy is a conserved catabolic process in which intracellular components such as protein/DNA aggregates and abnormal organelles are degraded in specific lysosomes. In cancer, autophagy plays a controversial role, acting in different conditions as both a tumor suppressor and a tumor-promoting mechanism. Experimental evidence shows that deep interrelations exist between EMT and autophagy-related pathways. Although this interplay has already been analyzed in previous studies, understanding mechanisms and the translational implications of autophagy/EMT need further study. The role of autophagy in EMT is not limited to morphological changes, but activation of autophagy could be important to DNA repair/damage system, cell adhesion molecules, and cell proliferation and differentiation processes. Based on this, both autophagy and EMT and related pathways are now considered as targets for cancer therapy. In this review article, the contribution of autophagy to EMT and progression of cancer is discussed. This article also describes the multiple connections between EMT and autophagy and their implication in cancer treatment.
Collapse
Affiliation(s)
- Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy;
- National Institute for Infectious Diseases “Lazzaro Spallanzani”, I.R.C.C.S., 00149 Rome, Italy
| | - Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modares University, Tehran P.O. Box 14115-331, Iran;
| | - Maryam Adelipour
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran;
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye;
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain;
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye;
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modares University, Tehran P.O. Box 14115-331, Iran;
| |
Collapse
|
11
|
Kim D, Kim DH. Subcellular mechano-regulation of cell migration in confined extracellular microenvironment. BIOPHYSICS REVIEWS 2023; 4:041305. [PMID: 38505424 PMCID: PMC10903498 DOI: 10.1063/5.0185377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/01/2023] [Indexed: 03/21/2024]
Abstract
Cell migration is a highly coordinated cellular event that determines diverse physiological and pathological processes in which the continuous interaction of a migrating cell with neighboring cells or the extracellular matrix is regulated by the physical setting of the extracellular microenvironment. In confined spaces, cell migration occurs differently compared to unconfined open spaces owing to the additional forces that limit cell motility, which create a driving bias for cells to invade the confined space, resulting in a distinct cell motility process compared to what is expected in open spaces. Moreover, cells in confined environments can be subjected to elevated mechanical compression, which causes physical stimuli and activates the damage repair cycle in the cell, including the DNA in the nucleus. Although cells have a self-restoring system to repair damage from the cell membrane to the genetic components of the nucleus, this process may result in genetic and/or epigenetic alterations that can increase the risk of the progression of diverse diseases, such as cancer and immune disorders. Furthermore, there has been a shift in the paradigm of bioengineering from the development of new biomaterials to controlling biophysical cues and fine-tuning cell behaviors to cure damaged/diseased tissues. The external physical cues perceived by cells are transduced along the mechanosensitive machinery, which is further channeled into the nucleus through subcellular molecular linkages of the nucleoskeleton and cytoskeleton or the biochemical translocation of transcription factors. Thus, external cues can directly or indirectly regulate genetic transcriptional processes and nuclear mechanics, ultimately determining cell fate. In this review, we discuss the importance of the biophysical cues, response mechanisms, and mechanical models of cell migration in confined environments. We also discuss the effect of force-dependent deformation of subcellular components, specifically focusing on subnuclear organelles, such as nuclear membranes and chromosomal organization. This review will provide a biophysical perspective on cancer progression and metastasis as well as abnormal cellular proliferation.
Collapse
Affiliation(s)
- Daesan Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | | |
Collapse
|
12
|
Saito J, Zao H, Wu L, Iwasaki M, Sun Q, Hu C, Ishikawa M, Hirota K, Ma D. "Anti-cancer" effect of ketamine in comparison with MK801 on neuroglioma and lung cancer cells. Eur J Pharmacol 2023; 945:175580. [PMID: 36758782 DOI: 10.1016/j.ejphar.2023.175580] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
Ketamine, a N-methyl-D-aspartate (NMDA) receptor antagonist, is commonly used to induce anaesthesia during cancer surgery and relieve neuropathic and cancer pain. This study was conducted to assess whether ketamine has any inhibiting effects on neuroglioma (H4) and lung cancer cells (A549) in vitro. The cultured H4 and A549 cells were treated with ketamine and MK801 (0.1, 1, 10, 100, or 1000 μM) for 24 h. The expressions of glutamate receptors on both types of cancer cells were assessed with qRT-PCR. In addition, cell proliferation and migration were assessed with cell counting Kit-8 and wound healing assays. Cyclin D1, matrix metalloproteinase 9 (MMP9), phosphorylation of extracellular signal-regulated kinase (pERK), and cleaved-caspase-3 expression together with reactive oxygen species (ROS) were also assessed with Western blot, immunostaining, and/or flowcytometry. NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors were expressed on both H4 and A549 cells. Ketamine inhibited cancer cell proliferation and migration in a dose-dependent manner by suppressing the cell cycle and inducing apoptosis. Ketamine decreased cyclin D1, pERK, and MMP9 expression. In addition, ketamine increased ROS and cleaved caspase-3 expression and induced apoptosis. The anti-cancer effect of ketamine was more pronounced in A549 cells when compared with H4 cells. MK801 showed similar effects to those of ketamine. Ketamine suppressed cell proliferation and migration in both neuroglioma and lung cancer cells, likely through the antagonization of NMDA receptors.
Collapse
Affiliation(s)
- Junichi Saito
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK; Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | - Hailin Zao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | - Lingzhi Wu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | - Masae Iwasaki
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK; Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | - Qizhe Sun
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | - Cong Hu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | - Masashi Ishikawa
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK; Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | - Kazuyoshi Hirota
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK; National Clinical Research Center for Child Health, Hangzhou, China.
| | | |
Collapse
|
13
|
Jacksi M, Schad E, Buday L, Tantos A. Absence of Scaffold Protein Tks4 Disrupts Several Signaling Pathways in Colon Cancer Cells. Int J Mol Sci 2023; 24:ijms24021310. [PMID: 36674824 PMCID: PMC9861885 DOI: 10.3390/ijms24021310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
Tks4 is a large scaffold protein in the EGFR signal transduction pathway that is involved in several cellular processes, such as cellular motility, reactive oxygen species-dependent processes, and embryonic development. It is also implicated in a rare developmental disorder, Frank-ter Haar syndrome. Loss of Tks4 resulted in the induction of an EMT-like process, with increased motility and overexpression of EMT markers in colorectal carcinoma cells. In this work, we explored the broader effects of deletion of Tks4 on the gene expression pattern of HCT116 colorectal carcinoma cells by transcriptome sequencing of wild-type and Tks4 knockout (KO) cells. We identified several protein coding genes with altered mRNA levels in the Tks4 KO cell line, as well as a set of long non-coding RNAs, and confirmed these changes with quantitative PCR on a selected set of genes. Our results show a significant perturbation of gene expression upon the deletion of Tks4, suggesting the involvement of different signal transduction pathways over the well-known EGFR signaling.
Collapse
Affiliation(s)
- Mevan Jacksi
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, 1117 Budapest, Hungary
| | - Eva Schad
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - László Buday
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary
- Department of Molecular Biology, Semmelweis University Medical School, 1094 Budapest, Hungary
| | - Agnes Tantos
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary
- Correspondence:
| |
Collapse
|
14
|
Ballestri M, Marras E, Caruso E, Bolognese F, Malacarne MC, Martella E, Tubertini M, Gariboldi MB, Varchi G. Free and Poly-Methyl-Methacrylate-Bounded BODIPYs: Photodynamic and Antimigratory Effects in 2D and 3D Cancer Models. Cancers (Basel) 2022; 15:cancers15010092. [PMID: 36612089 PMCID: PMC9817850 DOI: 10.3390/cancers15010092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Several limitations, including dark toxicity, reduced tumor tissue selectivity, low photostability and poor biocompatibility hamper the clinical use of Photodynamic therapy (PDT) in cancer treatment. To overcome these limitations, new PSs have been synthetized, and often combined with drug delivery systems, to improve selectivity and reduce toxicity. In this context, BODIPYs (4,4-difluoro-4-bora-3a,4a-diaza-s-indacene) have recently emerged as promising and easy-to-handle scaffolds for the preparation of effective PDT antitumor agents. In this study, the anticancer photodynamic effect of newly prepared negatively charged polymethyl methacrylate (nPMMA)-bounded BODIPYs (3@nPMMA and 6@nPMMA) was evaluated on a panel of 2D- and 3D-cultured cancer cell lines and compared with free BODIPYs. In particular, the effect on cell viability was evaluated, along with their ability to accumulate into the cells, induce apoptotic and/or necrotic cell death, and inhibit cellular migration. Our results indicated that 3@nPMMA and 6@nPMMA reduce cancer cell viability in 3D models of HC116 and MCF7 cells more effectively than the corresponding free compounds. Importantly, we demonstrated that MDA-MB231 and SKOV3 cell migration ability was significantly impaired by the PDT treatment mediated by 3@nPMMA and 6@nPMMA nanoparticles, likely indicating the capability of this approach to reduce metastatic tumor potential.
Collapse
Affiliation(s)
- Marco Ballestri
- Institute for the Organic Synthesis and Photoreactivity, Italian National Research Council, 40129 Bologna, Italy
| | - Emanuela Marras
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Enrico Caruso
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Fabrizio Bolognese
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Miryam Chiara Malacarne
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Elisa Martella
- Institute for the Organic Synthesis and Photoreactivity, Italian National Research Council, 40129 Bologna, Italy
| | - Matilde Tubertini
- Institute for the Organic Synthesis and Photoreactivity, Italian National Research Council, 40129 Bologna, Italy
| | - Marzia Bruna Gariboldi
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
- Correspondence: (M.B.G.); (G.V.); Tel.: +39-033-133-9418 (M.B.C.); +39-051-639-8283 (G.V.)
| | - Greta Varchi
- Institute for the Organic Synthesis and Photoreactivity, Italian National Research Council, 40129 Bologna, Italy
- Correspondence: (M.B.G.); (G.V.); Tel.: +39-033-133-9418 (M.B.C.); +39-051-639-8283 (G.V.)
| |
Collapse
|
15
|
Doxycycline Hydrochloride Regulates Cytoskeletal Rearrangement and Epithelial-To-Mesenchymal Transition in Malignant Rhabdoid Tumour of the Kidney. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2760744. [DOI: 10.1155/2022/2760744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/15/2022]
Abstract
Objective. As a highly malignant tumour, malignant rhabdoid tumours of the kidney (MRTK) are prone to metastasis and invasion, while tumour metastasis and invasion are inseparable from matrix metalloproteinases (MMPs) and epithelial-mesenchymal transformation (EMT). Moreover, the key to EMT is remodelling of the cytoskeleton. Therefore, our study is aimed at investigating whether doxycycline hydrochloride (DCH), an inhibitor of MMPs, could reverse EMT in MRTK to exert an antitumour effect by regulating MMPs and the cytoskeleton. Methods. The existence of EMT in MRTK cells was verified by bioinformatics analysis, immunofluorescence, and western blotting (WB). In vitro, the proliferation, migration, and invasion abilities of G401 cells were examined by Cell Counting Kit-8 (CCK-8), scratch, and Transwell assays, respectively. The effect of DCH on tumour growth in tumour-bearing mice was explored in in vivo experiments, and the expression of MMP2 and MMP9 and EMT correlation indexes was measured by immunofluorescence and WB, and the changes in cytoskeletal F-actin and β-tubulin were measured by fluorescence. Results. The altered extracellular matrix (ECM) composition, EMT, and high expression of MMP2 and MMP9 existed in MRTK. DCH inhibited the proliferation, migration, and invasion of G401 cells in vitro. In vivo, DCH inhibited tumour growth in mice, downregulated the expression of MMP2 and MMP9, and partially reversed EMT. Alternatively, DCH resulted in cytoskeletal rearrangements of G401 cells. Conclusions. DCH, as an MMP inhibitor, is used for the first time in MRTK research, showing good antitumour effects by reversing EMT and potentially providing new therapeutic measures for MRTK treatment.
Collapse
|
16
|
Wang L, Liang J, Bi S, Li Y, Zhang W, Xiwen W, Liu Y, Liu H. Role of GLI1 in Hypoxia-Driven Endometrial Stromal Cell Migration and Invasion in Endometriosis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6890790. [PMID: 36285283 PMCID: PMC9588377 DOI: 10.1155/2022/6890790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/04/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022]
Abstract
Endometriosis (EMs) is a benign disease with the characteristics of invasion and migration, and its pathogenesis is related to hypoxia. The abnormal activation of glioma-associated oncogene homolog 1 (GLI1) plays an important role in the metastasis of multiple types of tumors. However, it is not clear whether GLI1 regulates the migration and invasion of endometrial stromal cells under hypoxic condition. Therefore, we use comprehensive analysis to explore the effects of hypoxic on GLI1 expression and their regulation on the pathogenesis of EMs. In this study, from immunohistochemistry, RT-qPCR, and western blot analysis, we discovered that the expression of hypoxia-induced factor-1α (HIF-1α) and GLI1 was significantly increased in eutopic and ectopic endometrium of patients with EMs. In human primary eutopic endometrial stromal cells (ESCs), hypoxia can increase the expression of HIF-1α and GLI1 in a time-dependent manner. And hypoxia could promote GLI1 expression in a HIF-1α-dependent manner. Moreover, data from transwell assays manifested that the migration and invasion ability of ESCs was significantly enhanced under hypoxia, and this effect could be reversed by silencing GLI1. Furthermore, the expression of MMP2 and MMP9 was also increased under hypoxia, while silencing GLI1 could reverse this event. In summary, our research verified that GLI1, which activated by hypoxia, may contribute to the migration and invasion of ESCs through the upregulation of MMP2 and MMP9 and can be a novel therapeutic target in EMs.
Collapse
Affiliation(s)
- Lili Wang
- Department of Obstetrics and Gynecology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaxin Liang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Siyi Bi
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yixuan Li
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Wang Xiwen
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yi Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hengwei Liu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| |
Collapse
|
17
|
Discovery of Simple Diacylhydrazine-Functionalized Cinnamic Acid Derivatives as Potential Microtubule Stabilizers. Int J Mol Sci 2022; 23:ijms232012365. [PMID: 36293224 PMCID: PMC9604255 DOI: 10.3390/ijms232012365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/21/2022] [Accepted: 10/05/2022] [Indexed: 11/04/2022] Open
Abstract
To develop novel microtubule-binding agents for cancer therapy, an array of N-cinnamoyl-N'-(substituted)acryloyl hydrazide derivatives were facilely synthesized through a two-step process. Initially, the antiproliferative activity of these title compounds was explored against A549, 98 PC-3 and HepG2 cancer cell lines. Notably, compound I23 exhibited the best antiproliferative activity against three cancer lines with IC50 values ranging from 3.36 to 5.99 μM and concurrently afforded a lower cytotoxicity towards the NRK-52E cells. Anticancer mechanism investigations suggested that the highly bioactive compound I23 could potentially promote the protofilament assembly of tubulin, thus eventually leading to the stagnation of the G2/M phase cell cycle of HepG2 cells. Moreover, compound I23 also disrupted cancer cell migration and significantly induced HepG2 cells apoptosis in a dosage-dependent manner. Additionally, the in silico analysis indicated that compound I23 exhibited an acceptable pharmacokinetic profile. Overall, these easily prepared N-cinnamoyl-N'-(substituted)acryloyl hydrazide derivatives could serve as potential microtubule-interacting agents, probably as novel microtubule-stabilizers.
Collapse
|
18
|
Mumu M, Das A, Emran TB, Mitra S, Islam F, Roy A, Karim MM, Das R, Park MN, Chandran D, Sharma R, Khandaker MU, Idris AM, Kim B. Fucoxanthin: A Promising Phytochemical on Diverse Pharmacological Targets. Front Pharmacol 2022; 13:929442. [PMID: 35983376 PMCID: PMC9379326 DOI: 10.3389/fphar.2022.929442] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
Fucoxanthin (FX) is a special carotenoid having an allenic bond in its structure. FX is extracted from a variety of algae and edible seaweeds. It has been proved to contain numerous health benefits and preventive effects against diseases like diabetes, obesity, liver cirrhosis, malignant cancer, etc. Thus, FX can be used as a potent source of both pharmacological and nutritional ingredient to prevent infectious diseases. In this review, we gathered the information regarding the current findings on antimicrobial, antioxidant, anti-inflammatory, skin protective, anti-obesity, antidiabetic, hepatoprotective, and other properties of FX including its bioavailability and stability characteristics. This review aims to assist further biochemical studies in order to develop further pharmaceutical assets and nutritional products in combination with FX and its various metabolites.
Collapse
Affiliation(s)
- Mumtaza Mumu
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- *Correspondence: Talha Bin Emran, ; Abubakr M. Idris, ; Bonglee Kim,
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Arpita Roy
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Md. Mobarak Karim
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Rajib Das
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Moon Nyeo Park
- Department of Pathology College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway, Malaysia
| | - Abubakr M. Idris
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
- *Correspondence: Talha Bin Emran, ; Abubakr M. Idris, ; Bonglee Kim,
| | - Bonglee Kim
- Department of Pathology College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- *Correspondence: Talha Bin Emran, ; Abubakr M. Idris, ; Bonglee Kim,
| |
Collapse
|
19
|
Lindgren M, Rask G, Jonsson J, Berglund A, Lundin C, Jonsson P, Ljuslinder I, Nyström H. Type IV Collagen in Human Colorectal Liver Metastases—Cellular Origin and a Circulating Biomarker. Cancers (Basel) 2022; 14:cancers14143396. [PMID: 35884455 PMCID: PMC9325127 DOI: 10.3390/cancers14143396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Patients with colorectal liver metastases (CLM) can be cured through surgery if metastases are detected early in disease progression. Today, CLM diagnosis relies heavily on diagnostic imaging, and cheap, non-invasive, and efficiently measurable biomarkers are needed. Circulating type IV collagen (COL IV) is a potential biomarker for detecting CLM. Patients with CLM show elevated circulating levels of COL IV and increased tissue expression of COL IV in CLM tissue, which could result from enhanced production and degradation of COL IV. This study aimed to establish the cellular source behind enhanced COL IV levels, which is helpful in the evaluation of the biomarker potential of COL IV. We show that fibroblasts express COL IV both in vitro and in the stromal tissue of CLM. We also found that CLM tissue expresses COL IV-degrading proteases. Lastly, CLM patients have higher circulating COL IV levels than healthy controls. Abstract Circulating type IV collagen (cCOL IV) is a potential biomarker for patients with colorectal liver metastases (CLM) who present with elevated levels of COL IV in both CLM tissue and circulation. This study aimed to establish the cellular origin of elevated levels of COL IV and analyze circulating COL IV in CLM patients. The cellular source was established through in situ hybridization, immunohistochemical staining, and morphological evaluation. Cellular expression in vitro was assessed by immunofluorescence. Tissue expression of COL IV-degrading matrix metalloproteinases (MMPs)-2, -7, -9, and -13 was studied with immunohistochemical staining. Plasma levels of COL IV in CLM patients and healthy controls were analyzed with ELISA. This study shows that cancer-associated fibroblasts (CAFs) express COL IV in the stroma of CLM and that COL IV is expressed in vitro by fibroblasts but not by tumor cells. MMP-2, -7, -9, and -13 are expressed in CLM tissue, mainly by hepatocytes and immune cells, and circulating COL IV is significantly elevated in CLM patients compared with healthy controls. Our study shows that stromal cells, not tumor cells, produce COL IV in CLM, and that circulating COL IV is elevated in patients with CLM.
Collapse
Affiliation(s)
- Moa Lindgren
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, SE-901 85 Umeå, Sweden; (G.R.); (J.J.); (A.B.); (C.L.); (H.N.)
- Correspondence:
| | - Gunilla Rask
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, SE-901 85 Umeå, Sweden; (G.R.); (J.J.); (A.B.); (C.L.); (H.N.)
- Department of Medical Biosciences/Pathology, Umeå University, SE-901 87 Umeå, Sweden
| | - Josefin Jonsson
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, SE-901 85 Umeå, Sweden; (G.R.); (J.J.); (A.B.); (C.L.); (H.N.)
| | - Anette Berglund
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, SE-901 85 Umeå, Sweden; (G.R.); (J.J.); (A.B.); (C.L.); (H.N.)
| | - Christina Lundin
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, SE-901 85 Umeå, Sweden; (G.R.); (J.J.); (A.B.); (C.L.); (H.N.)
| | - Pär Jonsson
- Department of Chemistry, Umeå University, SE-907 36 Umeå, Sweden;
| | - Ingrid Ljuslinder
- Department of Radiation Sciences/Oncology, Umeå University, SE-901 87 Umeå, Sweden;
| | - Hanna Nyström
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, SE-901 85 Umeå, Sweden; (G.R.); (J.J.); (A.B.); (C.L.); (H.N.)
- Wallenberg Centre for Molecular Medicine, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
20
|
Li S, Yue H, Wang S, Li X, Wang X, Guo P, Ma G, Wei W. Advances of bacteria-based delivery systems for modulating tumor microenvironment. Adv Drug Deliv Rev 2022; 188:114444. [PMID: 35817215 DOI: 10.1016/j.addr.2022.114444] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 12/13/2022]
Abstract
The components and hospitable properties of tumor microenvironment (TME) are associated with tumor progression. Recently, TME modulating vectors and strategies have garnished significant attention in cancer therapy. Although a pilot work has reviewed TME regulation via nanoparticle-based delivery systems, there is no systematical review that summarizes the natural bacteria-based anti-tumor system to modulate TME. In this review, we conclude the strategies of bacterial carriers (including whole bacteria, bacterial skeleton and bacterial components) to regulate TME from the perspective of TME components and hospitable properties, and the clinical trials of bacteria-mediated cancer therapy. Current challenges and future prospects for the design of bacteria-based carriers are also proposed that provide critical insights into this natural delivery system and related translation from the bench to the clinic.
Collapse
Affiliation(s)
- Shuping Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Hua Yue
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xin Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xiaojun Wang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Peilin Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
21
|
Alba J, Barcia R, Gutiérrez-Berzal J, Ramos-Martínez JI. Could inhibition of metalloproteinases be used to block the process of metastasis? Cell Biochem Funct 2022; 40:600-607. [PMID: 35789101 PMCID: PMC9544369 DOI: 10.1002/cbf.3730] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 12/14/2022]
Abstract
Metastasis is a multisequential process that allows tumor cells to migrate to tissues distant from the primary tumor. Only a small number of cells escape from the primary tumor; however, the metastases generated are responsible for more than 90% of cancer deaths. Many metastatic processes initially require the total or partial start‐up of a program for the transformation of tumor epithelial cells into mesenchymal cells (EMT). The launching of the EMT program is stimulated by cytokines and other elements produced by the diverse types of cells composing the tumor stroma. In parallel, a process of destabilization of the extracellular matrix (ECM) takes place by means of the synthesis of proteases of the matrix metalloproteinases (MMPs) family. EMC degradation allows the exportation of some tumor cells as mesenchymal cells to the circulatory system and their subsequent implantation in a tissue distant from the primary tumor. The blocking of these both processes appears as a hypothetical stop point in the metastatic mechanism. The present review deals with the different options to achieve the inhibition of MMPs, focusing on MMP7 as a target given its involvement in the metastatic processes of a wide variety of tumors. The simultaneous implantation of the epithelial–mesenchymal program and the synthesis and activation of matrix metalloproteinases during the first phases of the metastasis process is known. The inhibition of proteases could constitute a possible blockage of the process. The review describes the evolution of the different inhibition mechanisms that could inform applicable therapeutic mechanisms for the paralysis of the metastatic process.
Collapse
Affiliation(s)
- Jesús Alba
- Histobiomol, Hospital POLUSA, Lugo, Spain
| | - Ramiro Barcia
- Faculty of Sciences, University of Santiago de Compostela, Lugo, Spain
| | | | - Juan I Ramos-Martínez
- Department of Biochemistry and Molecular Biology, School of Veterinary, University of Santiago de Compostela, Lugo, Spain
| |
Collapse
|
22
|
AlKahlout A, Fardoun M, Mesmar J, Abdallah R, Badran A, Nasser SA, Baydoun S, Kobeissy F, Shaito A, Iratni R, Muhammad K, Baydoun E, Eid AH. Origanum syriacum L. Attenuates the Malignant Phenotype of MDA-MB231 Breast Cancer Cells. Front Oncol 2022; 12:922196. [PMID: 35847867 PMCID: PMC9280492 DOI: 10.3389/fonc.2022.922196] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/25/2022] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is the leading cause of cancer-related deaths among women. Among breast cancer types, triple negative breast cancer (TNBC) is the most aggressive, and is resistant to hormonal and chemotherapeutic treatments. As such, alternative approaches that may provide some benefit in fighting this debilitating pathology are critically needed; hence the utilization of herbal medicine. Origanum syriacum L., one of the most regularly consumed plants in the Mediterranean region, exhibits antiproliferative effect on several cancer cell lines. However, whether this herb modulates the malignant phenotype of TNBC remains poorly investigated. Here, we show that in MDA-MB-231, a TNBC cell line, Origanum syriacum L. aqueous extract (OSE) inhibited cellular viability, induced autophagy determined by the accumulation of lipidized LC3 II, and triggered apoptosis. We also show that OSE significantly promoted homotypic cell-cell adhesion while it decreased cellular migration, adhesion to fibronectin, and invasion of MDA-MB-231 cells. This was supported by decreased activity of focal adhesion kinase (FAK), reduced α2 integrin expression, and downregulation of secreted PgE2, MMP2 and MMP-9, in OSE-treated cells. Finally, we also show that OSE significantly inhibited angiogenesis and downregulated the level of nitric oxide (NO) production. Our findings demonstrate the ability of OSE to attenuate the malignant phenotype of the MDA-MB-231 cells, thus presenting Origanum syriacum L. as a promising potential source for therapeutic compounds for TNBC.
Collapse
Affiliation(s)
| | - Manal Fardoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Joelle Mesmar
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Rola Abdallah
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Adnan Badran
- Department of Basic Sciences, University of Petra, Amman, Jordan
| | - Suzanne A. Nasser
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | - Serine Baydoun
- Department of Diagnostic Radiology, Cleveland Clinic, Cleveland, OH, United States
| | - Firas Kobeissy
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainseville, FL, United States
| | | | - Rabah Iratni
- Department of Biology, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Khalid Muhammad
- Department of Biology, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
23
|
Mahdieh Z, Cherne MD, Fredrikson JP, Sidar B, Sanchez HS, Chang CB, Bimczok D, Wilking JN. Granular Matrigel: restructuring a trusted extracellular matrix material for improved permeability. Biomed Mater 2022; 17:045020. [PMID: 35609584 DOI: 10.1088/1748-605x/ac7306] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/24/2022] [Indexed: 11/11/2022]
Abstract
Matrigel is a polymeric extracellular matrix material produced by mouse cancer cells. Over the past four decades, Matrigel has been shown to support a wide variety of two- and three-dimensional cell and tissue culture applications including organoids. Despite widespread use, transport of molecules, cells, and colloidal particles through Matrigel can be limited. These limitations restrict cell growth, viability, and function and limit Matrigel applications. A strategy to improve transport through a hydrogel without modifying the chemistry or composition of the gel is to physically restructure the material into microscopic microgels and then pack them together to form a porous material. These 'granular' hydrogels have been created using a variety of synthetic hydrogels, but granular hydrogels composed of Matrigel have not yet been reported. Here we present a drop-based microfluidics approach for structuring Matrigel into a three-dimensional, mesoporous material composed of packed Matrigel microgels, which we call granular Matrigel. We show that restructuring Matrigel in this manner enhances the transport of colloidal particles and human dendritic cells (DCs) through the gel while providing sufficient mechanical support for culture of human gastric organoids (HGOs) and co-culture of human DCs with HGOs.
Collapse
Affiliation(s)
- Zahra Mahdieh
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Michelle D Cherne
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States of America
| | - Jacob P Fredrikson
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Barkan Sidar
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Humberto S Sanchez
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Connie B Chang
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Diane Bimczok
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States of America
| | - James N Wilking
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| |
Collapse
|
24
|
Expression of Matrix Metalloproteinase-7 Predicts Poor Prognosis in Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2300979. [PMID: 35496040 PMCID: PMC9054443 DOI: 10.1155/2022/2300979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/31/2022] [Indexed: 11/17/2022]
Abstract
To date, no potential markers have been established for predicting prognosis in gastric cancer. Matrix metalloproteinase-7 (MMP-7) has been suggested as a prognostic marker in several cancers. In this study, we aimed to determine the expression of the MMP-7 protein and its polymorphisms in gastric cancer tissues. The association between MMP-7 expression level and clinicopathological characteristics was also evaluated. MMP-7 protein expression and its polymorphisms were investigated in a total of 400 patients using immunohistochemistry and TaqMan SNP genotyping assays. The correlation of MMP-7 expression with clinicopathological characteristics, including tumor location, tumor size, histologic type, lymphatic invasion, vascular invasion, pathological T stage, pathological TNM stage, residual tumor, and CEA level, was investigated. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using a multivariate Cox proportional hazards regression model. MMP-7 expression was found in 283 of 400 (70.75%) gastric cancer tissues. Expression of MMP-7 was significantly associated with poor clinicopathological characteristics, including vascular invasion (
,
,
), lymphatic invasion (
,
,
), undifferentiated histologic type (
; 95% CI, 1.31–4.52;
), higher TNM stage (stage IV) (
,
,
), and high CEA level (
,
,
). We further observed a significant association of the variant genotype; gastric cancer patients carrying GG of MMP-7 (-181A/G; rs11568818) had a greater increased risk of MMP-7 expression than did wild-type (WT) carriers (AG: odds ratio
;
;
and GG:
;
;
). These findings suggest that MMP-7 expression can be used to predict the prognosis of gastric cancer patients.
Collapse
|
25
|
Yang F, Ye R, Ma C, Wang Y, Wang Y, Chen J, Yang J, Höfer J, Zhu Y, Xiao L, Zhang J, Xu Y. Toxicity evaluation, toxin screening and its intervention of the jellyfish Phacellophora camtschatica based on a combined transcriptome-proteome analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 233:113315. [PMID: 35189521 DOI: 10.1016/j.ecoenv.2022.113315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The application of multi-omics technologies provides a new perspective to solve three main problems including species identification, toxin screening and effective antagonist conformation in the studies of marine toxic jellyfish. METHODS A series of transcriptome-proteome based analysis accompanied with toxicity evaluations were performed for the ornamental jellyfish Phacellophora camtschatica. RESULTS Through combined morphological observation and Cytochrome c oxidase subunit Ⅰ (CO1) molecular alignment, the sample jellyfish was identified as P. camtschatica. A total of 25,747 unigenes and 3058 proteins were obtained from the successfully constructed transcriptome and proteome, in which 6869 (26.68%) and 6618 (25.70%) unigenes, as well as 2536 (82.93%) and 2844 (93.00%) proteins were annotated against the databases of Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), respectively. The jellyfish displayed obvious in vivo lethal effects with significant increases of multi-organ functional indexes as well as in vitro activities. Total of 62 toxins from 120 toxin-related unigenes were screened including 16 metalloproteases, 11 phospholipases and others. Moreover, 11 toxins were further screened by using the erythrocyte model, where the zinc metalloproteinase nas-15-like (1) was the most abundant. Finally, Diltiazem greatly improved the survival rate while EDTA slightly prolonged the survival time in ICR mice. CONCLUSION P. camtschatica is a poisonous jellyfish with diversified toxic components, in which metalloproteinase probably plays an important role in toxicities, and excessive Ca2+ entry may be the main mechanism of systemic lethal toxicity.
Collapse
Affiliation(s)
- Fengling Yang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Ruiwei Ye
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Chaoqun Ma
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Rd, Shanghai 200433, China.
| | - Yichao Wang
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Department of Clinical Laboratory, Taizhou Central Hospital, Taizhou 318000, China.
| | - Yi Wang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Jianmei Chen
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Jishun Yang
- Medical Insurance Center, Navy Medical Center, Navy Medical Center of PLA, Shanghai 200050, China.
| | - Juan Höfer
- Escuela de Ciencias del Mar, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.
| | - Yina Zhu
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Liang Xiao
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Jing Zhang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China.
| | - Yinghe Xu
- Department of Intensive Care Unit, Taizhou Central Hospital, Taizhou 318000, China.
| |
Collapse
|
26
|
Shen J, Han L, Xue Y, Li C, Jia H, Zhu K. Ropivacaine Inhibits Lung Cancer Cell Malignancy Through Downregulation of Cellular Signaling Including HIF-1α In Vitro. Front Pharmacol 2022; 12:806954. [PMID: 35280249 PMCID: PMC8905340 DOI: 10.3389/fphar.2021.806954] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/28/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Ropivacaine is widely used to induce regional anesthesia during lung cancer surgery. Previous studies reported that amide-linked local anesthetics, e.g., ropivacaine, affected the biological behavior of lung adenocarcinoma cells, but the conclusion is controversial and warrants further study. This study set out to investigate the biological effects of ropivacaine on cultured lung cancer cells and underlying mechanisms. Methods: Lung cancer cell lines (A549 and H1299) were cultured and then treated with or without ropivacaine (0.5, 1, and 2 mM) for 48 or 72 h. Their proliferation, migration, and invasion together with cell death and molecules including hypoxia inducible factor (HIF)-1α, VEGF, matrix metalloproteinase (MMP)-1, MMP-2, and MMP-9 expression associated with these changes were determined. Results: Ropivacaine significantly inhibited proliferation and migration, invasion, and cell death in a concentration-dependent manner in both cell lines. Ropivacaine also promoted cell death and induced a concentration- and time-dependent cell arrest towards the G0/G1 phase. Expression of VEGF, MMP-1, MMP-2, MMP-9, and HIF-1α in both cell lines was also inhibited by ropivacaine in a concentration-related manner. Conclusion: Our data indicated that ropivacaine inhibited lung cancer cell malignancy, which may be associated with downregulation of cell-survival-associated cellular molecules. The translational value of the current work is subjected to further study.
Collapse
Affiliation(s)
- Junmei Shen
- Department of Anesthesiology, The Forth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lina Han
- Department of Blood Transfusion, The Forth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yongxian Xue
- Scientific Research Center, The Forth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chao Li
- Department of Anesthesiology, The Forth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huiqun Jia
- Department of Anesthesiology, The Forth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Kangsheng Zhu
- Department of Anesthesiology, The Forth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
27
|
Binlateh T, Uppatcha N, Thepchai J, Pleungtuk Y, Noisa P, Hutamekalin P, Jitprasertwong P. Cordycepin attenuates migration and invasion of HSC-4 oral squamous carcinoma cells through autophagy-dependent FAK/Akt and MMP2/MMP9 suppression. J Dent Sci 2022; 17:1677-1688. [PMID: 36299321 PMCID: PMC9588793 DOI: 10.1016/j.jds.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/04/2022] [Indexed: 10/29/2022] Open
|
28
|
TIMP-2 regulates 5-Fu resistance via the ERK/MAPK signaling pathway in colorectal cancer. Aging (Albany NY) 2022; 14:297-315. [PMID: 35022331 PMCID: PMC8791226 DOI: 10.18632/aging.203793] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/03/2021] [Indexed: 12/24/2022]
Abstract
5-Fluorouracil (5-Fu) is the first-line chemotherapeutic option for colorectal cancer. However, its efficacy is inhibited by drug resistance. Cytokines play an important role in tumor drug resistance, even though their mechanisms are largely unknown. Using a cytokine array, we established that tissue inhibitor metalloproteinase 2 (TIMP-2) is highly expressed in 5-Fu resistant colorectal cancer patients. Analysis of samples from 84 patients showed that elevated TIMP-2 expression levels in colorectal patients were correlated with poor prognostic outcomes. In a 5-Fu-resistant patient-derived xenograft (PDX) model, TIMP-2 was also found to be highly expressed. We established an autocrine mechanism through which elevated TIMP-2 protein levels sustained colorectal cancer cell resistance to 5-Fu by constitutively activating the ERK/MAPK signaling pathway. Inhibition of TIMP-2 using an anti-TIMP-2 antibody or ERK/MAPK inhibition by U0126 suppressed TIMP-2 mediated 5-Fu-resistance in CRC patients. In conclusion, a novel TIMP-2-ERK/MAPK mediated 5-Fu resistance mechanism is involved in colorectal cancer. Therefore, targeting TIMP-2 or ERK/MAPK may provide a new strategy to overcome 5-Fu resistance in colorectal cancer chemotherapy.
Collapse
|
29
|
Geng L, Wang Z, Tian Y. Down-regulation of ZNF252P-AS1 alleviates ovarian cancer progression by binding miR-324-3p to downregulate LY6K. J Ovarian Res 2022; 15:1. [PMID: 34980214 PMCID: PMC8725409 DOI: 10.1186/s13048-021-00933-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 12/06/2021] [Indexed: 11/29/2022] Open
Abstract
Background Ovarian cancer is a common gynecological malignant disease in women. Our work aimed to study the specific functions of ZNF252P antisense RNA 1 (ZNF252P-AS1) in ovarian cancer. Methods ZNF252P-AS1, miR-324-3p, and lymphocyte antigen 6 family member K (LY6K) expression were analyzed by bioinformatics tools in ovarian cancer tissues and was quantified by qRT-PCR in ovarian cancer cells. The effect of ZNF252P-AS1 knockdown, miR-324-3p suppression, and LY6K over-expression on apoptosis, cell viability, invasion, migration, and epithelial to mesenchymal transition (EMT) was determined in vitro by using colony formation and EdU assays, flow cytometry, transwell assay, and Western blot. The interactions between ZNF252P-AS1 and miR-324-3p and between miR-324-3p and LY6K were validated by luciferase assays. The effects of restraining ZNF252P-AS1 in vivo were studied using BALB/c male nude mice. Results ZNF252P-AS1 and LY6K levels were up-regulated, while miR-324-3p was declined in ovarian cancer tissues and cells. ZNF252P-AS1 knockdown reduced ovarian cancer cell proliferation, invasion, migration, and EMT, whereas promoted its apoptosis. Besides, ZNF252P-AS1 interacted with miR-324-3p and reversely regulated its level, and miR-324-3p was directly bound to LY6K and negatively regulated its expression. Moreover, ZNF252P-AS1 knockdown reversed the effect of miR-324-3p on cancer cell apoptosis, growth, migration, invasion, and EMT. Similar results were discovered in the rescue experiments between miR-324-3p and LY6K. Additionally, mouse models in vivo experiments further validated that ZNF252P-AS1 knockdown distinctly inhibited tumor growth. Conclusion ZNF252P-AS1 mediated miR-324-3p/LY6K signaling to facilitate progression of ovarian cancer.
Collapse
Affiliation(s)
- Li Geng
- Department of Pathology, Jinan City People's Hospital, Shandong First Medical University, Jinan, Shandong, 271100, P.R. China
| | - Zhongqiu Wang
- Department of Pediatric Surgery, Jinan City People's Hospital, Shandong First Medical University, Jinan, Shandong, 271100, P.R. China
| | - Yongju Tian
- Department of Gynecology, Yantaishan Hospital, Yantai, Shandong, 264001, P.R. China.
| |
Collapse
|
30
|
He L, Zhong Z, Chen M, Liang Q, Wang Y, Tan W. Current Advances in Coptidis Rhizoma for Gastrointestinal and Other Cancers. Front Pharmacol 2022; 12:775084. [PMID: 35046810 PMCID: PMC8762280 DOI: 10.3389/fphar.2021.775084] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/08/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is a serious disease with an increasing number of reported cases and high mortality worldwide. Gastrointestinal cancer defines a group of cancers in the digestive system, e.g., liver cancer, colorectal cancer, and gastric cancer. Coptidis Rhizoma (C. Rhizoma; Huanglian, in Chinese) is a classical Chinese medicinal botanical drug for the treatment of gastrointestinal disorders and has been shown to have a wide variety of pharmacological activity, including antifungal, antivirus, anticancer, antidiabetic, hypoglycemic, and cardioprotective effects. Recent studies on C. Rhizoma present significant progress on its anticancer effects and the corresponding mechanisms as well as its clinical applications. Herein, keywords related to C. Rhizoma, cancer, gastrointestinal cancer, and omics were searched in PubMed and the Web of Science databases, and more than three hundred recent publications were reviewed and discussed. C. Rhizoma extract along with its main components, berberine, palmatine, coptisine, magnoflorine, jatrorrhizine, epiberberine, oxyepiberberine, oxyberberine, dihydroberberine, columbamine, limonin, and derivatives, are reviewed. We describe novel and classic anticancer mechanisms from various perspectives of pharmacology, pharmaceutical chemistry, and pharmaceutics. Researchers have transformed the chemical structures and drug delivery systems of these components to obtain better efficacy and bioavailability of C. Rhizoma. Furthermore, C. Rhizoma in combination with other drugs and their clinical application are also summarized. Taken together, C. Rhizoma has broad prospects as a potential adjuvant candidate against cancers, making it reasonable to conduct additional preclinical studies and clinical trials in gastrointestinal cancer in the future.
Collapse
Affiliation(s)
- Luying He
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Zhangfeng Zhong
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- *Correspondence: Zhangfeng Zhong, ; Yitao Wang, ; Wen Tan,
| | - Man Chen
- Oncology Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qilian Liang
- Oncology Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yitao Wang
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- *Correspondence: Zhangfeng Zhong, ; Yitao Wang, ; Wen Tan,
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, China
- *Correspondence: Zhangfeng Zhong, ; Yitao Wang, ; Wen Tan,
| |
Collapse
|
31
|
Gouda G, Gupta MK, Donde R, Behera L, Vadde R. Tumor microenvironment in heptocellular carcinoma. THERANOSTICS AND PRECISION MEDICINE FOR THE MANAGEMENT OF HEPATOCELLULAR CARCINOMA 2022:109-124. [DOI: 10.1016/b978-0-323-98806-3.00007-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
|
32
|
Shigeta S, Watanabe Y, Suzuki F, Nagase S, Shibuya Y, Ishibashi M, Nagai T, Shiga N, Toyoshima M, Tokunaga H, Shimada M, Yaegashi N. MicroRNA-152 Regulates Endometrial Serous Carcinoma Cell Motility by Suppressing Matrix Metalloproteinase 10 Expression. TOHOKU J EXP MED 2022; 256:249-258. [DOI: 10.1620/tjem.256.249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Shogo Shigeta
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Yoh Watanabe
- Division of Obstetrics and Gynecology, Tohoku Medical and Pharmaceutical University
| | - Fumihiko Suzuki
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Satoru Nagase
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine
| | - Yusuke Shibuya
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Masumi Ishibashi
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Tomoyuki Nagai
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Naomi Shiga
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | | | - Hideki Tokunaga
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Muneaki Shimada
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine
| |
Collapse
|
33
|
Wang L, Chen X, Wang L, Wang S, Li W, Liu Y, Zhang J. Knockdown of ST6Gal-I expression in human hepatocellular carcinoma cells inhibits their exosome-mediated proliferation- and migration-promoting effects. IUBMB Life 2021; 73:1378-1391. [PMID: 34559939 DOI: 10.1002/iub.2562] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/07/2021] [Accepted: 09/16/2021] [Indexed: 01/15/2023]
Abstract
Abnormal sialylation is a distinctive feature of human hepatocellular carcinoma (HCC) and is closely related to its malignant properties. Exosomes have characteristic protein and lipid composition; however, the results concerning glycoprotein composition and glycosylation are scarce. In this study, liquid chromatography-tandem mass spectrometry (LC-MS/MS) identified multiple microvesicle-related sialylated proteins including CD63, a classic marker of exosomes. The silencing of α2,6-sialyltransferase I (ST6Gal-I) significantly reduced the levels of α2,6-sialylated glycoconjugates on CD63 and the surface of HCC-derived exosomes (HCC-exo). And surface glycoconjugates play important roles in exosomes biogenesis and in their interaction with other cells. Compared to exosomes derived from naive HCC cells, α2,6-sialylation degradation abolished both the proliferation-promoting and migration-promoting effects of HCC-exo. Further analysis revealed that the Akt/GSK-3β or JNK1/2 signaling mediates HCC-exo-mediated proliferation in HCC cells, while ST6Gal-I silencing deactivated this pathway. These findings suggest that a loss of α2,6-sialylation decreases HCC progression through the loss of cancer cell-derived exosomes; furthermore, it opens novel perspectives to further explore the functional role of glycans in the biology of exosomes.
Collapse
Affiliation(s)
- Liping Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Xixi Chen
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Lingyan Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, China
| | - Wenli Li
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Yubo Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Jianing Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| |
Collapse
|
34
|
Mohammed S, Shamseddine AA, Newcomb B, Chavez RS, Panzner TD, Lee AH, Canals D, Okeoma CM, Clarke CJ, Hannun YA. Sublethal doxorubicin promotes migration and invasion of breast cancer cells: role of Src Family non-receptor tyrosine kinases. Breast Cancer Res 2021; 23:76. [PMID: 34315513 PMCID: PMC8317414 DOI: 10.1186/s13058-021-01452-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 07/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Doxorubicin (Dox) is a widely used chemotherapy, but its effectiveness is limited by dose-dependent side effects. Although lower Dox doses reduce this risk, studies have reported higher recurrence of local disease with no improvement in survival rate in patients receiving low doses of Dox. To effectively mitigate this, a better understanding of the adverse effects of suboptimal Dox doses is needed. METHODS Effects of sublethal dose of Dox on phenotypic changes were assessed with light and confocal microscopy. Migratory and invasive behavior were assessed by wound healing and transwell migration assays. MTT and LDH release assays were used to analyze cell growth and cytotoxicity. Flow cytometry was employed to detect cell surface markers of cancer stem cell population. Expression and activity of matrix metalloproteinases were probed with qRT-PCR and zymogen assay. To identify pathways affected by sublethal dose of Dox, exploratory RNAseq was performed and results were verified by qRT-PCR in multiple cell lines (MCF7, ZR75-1 and U-2OS). Regulation of Src Family kinases (SFK) by key players in DNA damage response was assessed by siRNA knockdown along with western blot and qRT-PCR. Dasatinib and siRNA for Fyn and Yes was employed to inhibit SFKs and verify their role in increased migration and invasion in MCF7 cells treated with sublethal doses of Dox. RESULTS The results show that sublethal Dox treatment leads to increased migration and invasion in otherwise non-invasive MCF7 breast cancer cells. Mechanistically, these effects were independent of the epithelial mesenchymal transition, were not due to increased cancer stem cell population, and were not observed with other chemotherapies. Instead, sublethal Dox induces expression of multiple SFK-including Fyn, Yes, and Src-partly in a p53 and ATR-dependent manner. These effects were validated in multiple cell lines. Functionally, inhibiting SFKs with Dasatinib and specific downregulation of Fyn suppressed Dox-induced migration and invasion of MCF7 cells. CONCLUSIONS Overall, this study demonstrates that sublethal doses of Dox activate a pro-invasive, pro-migration program in cancer cells. Furthermore, by identifying SFKs as key mediators of these effects, our results define a potential therapeutic strategy to mitigate local invasion through co-treatment with Dasatinib.
Collapse
Affiliation(s)
- Samia Mohammed
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794- 8430, USA
- Stony Brook University Cancer Center, MART Level 9, Stony Brook University, Stony Brook, NY, 11794-8430, USA
- Department of Medicine, Stony Brook University, Health Science Center, Hospital Pavilion Level 5, Stony Brook, NY, 11794-8430, USA
| | - Achraf A Shamseddine
- Department of Medicine, Stony Brook University, Health Science Center, Hospital Pavilion Level 5, Stony Brook, NY, 11794-8430, USA
| | - Benjamin Newcomb
- Department of Medicine, Stony Brook University, Health Science Center, Hospital Pavilion Level 5, Stony Brook, NY, 11794-8430, USA
| | - Ronald S Chavez
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794- 8430, USA
| | - Tyler D Panzner
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794-8430, USA
| | - Allen H Lee
- Stony Brook University Cancer Center, MART Level 9, Stony Brook University, Stony Brook, NY, 11794-8430, USA
- Department of Medicine, Stony Brook University, Health Science Center, Hospital Pavilion Level 5, Stony Brook, NY, 11794-8430, USA
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794-8430, USA
| | - Daniel Canals
- Stony Brook University Cancer Center, MART Level 9, Stony Brook University, Stony Brook, NY, 11794-8430, USA
- Department of Medicine, Stony Brook University, Health Science Center, Hospital Pavilion Level 5, Stony Brook, NY, 11794-8430, USA
| | - Chioma M Okeoma
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794-8430, USA
| | - Christopher J Clarke
- Stony Brook University Cancer Center, MART Level 9, Stony Brook University, Stony Brook, NY, 11794-8430, USA.
- Department of Medicine, Stony Brook University, Health Science Center, Hospital Pavilion Level 5, Stony Brook, NY, 11794-8430, USA.
| | - Yusuf A Hannun
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794- 8430, USA.
- Stony Brook University Cancer Center, MART Level 9, Stony Brook University, Stony Brook, NY, 11794-8430, USA.
- Department of Medicine, Stony Brook University, Health Science Center, Hospital Pavilion Level 5, Stony Brook, NY, 11794-8430, USA.
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794-8430, USA.
- The Northport Veterans Affairs Hospital, Northport, NY, 11768, USA.
| |
Collapse
|
35
|
The Application of Citrus folium in Breast Cancer and the Mechanism of Its Main Component Nobiletin: A Systematic Review. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2847466. [PMID: 34257674 PMCID: PMC8260297 DOI: 10.1155/2021/2847466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/28/2021] [Accepted: 06/19/2021] [Indexed: 02/07/2023]
Abstract
Citrus folium and its main ingredient nobiletin (NOB) have received widespread attention in recent years due to their antitumor effects. The antitumor effect of Citrus folium is related to the traditional use, mainly in its Chinese medicinal properties of soothing the liver and promoting qi, resolving phlegm, and dispelling stagnation. Some studies have proved that Citrus folium and NOB are more effective for triple-negative breast cancer (TNBC), which is related to the syndrome of stagnation of liver qi. From the perspective of modern biomedical research, NOB has anticancer effects. Its potential molecular mechanisms include inhibition of the cell cycle, induction of apoptosis, and inhibition of angiogenesis, invasion, and migration. Citrus folium and NOB can also reduce the side effects of chemotherapy drugs and reverse multidrug resistance (MDR). However, more research studies are needed to clarify the underlying mechanisms. The modern evidence of Citrus folium and NOB in breast cancer treatment has a strong connection with the traditional concepts and laws of applying Citrus folium in Chinese medicine (CM). As a low-toxic anticancer drug candidate, NOB and its structural changes, Citrus folium, and compound prescriptions will attract scientists to use advanced technologies such as genomics, proteomics, and metabolomics to study its potential anticancer effects and mechanisms. On the contrary, there are relatively few studies on the anticancer effects of Citrus folium and NOB in vivo. The clinical application of Citrus folium and NOB as new cancer treatment drugs requires in vivo verification and further anticancer mechanism research. This review aims to provide reference for the treatment of breast cancer by Chinese medicine.
Collapse
|
36
|
Giorello MB, Borzone FR, Labovsky V, Piccioni FV, Chasseing NA. Cancer-Associated Fibroblasts in the Breast Tumor Microenvironment. J Mammary Gland Biol Neoplasia 2021; 26:135-155. [PMID: 33398516 DOI: 10.1007/s10911-020-09475-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Years of investigation have shed light on a theory in which breast tumor epithelial cells are under the effect of the stromal microenvironment. This review aims to discuss recent findings concerning the phenotypic and functional characteristics of cancer associated fibroblasts (CAFs) and their involvement in tumor evolution, as well as their potential implications for anti-cancer therapy. In this manuscript, we reviewed that CAFs play a fundamental role in initiation, growth, invasion, and metastasis of breast cancer, and also serve as biomarkers in the clinical diagnosis, therapy, and prognosis of this disease.
Collapse
Affiliation(s)
- María Belén Giorello
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Vivian Labovsky
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Valeria Piccioni
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos (IBYME) y Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
37
|
CC Chemokine Ligand 7 Derived from Cancer-Stimulated Macrophages Promotes Ovarian Cancer Cell Invasion. Cancers (Basel) 2021; 13:cancers13112745. [PMID: 34206004 PMCID: PMC8198020 DOI: 10.3390/cancers13112745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022] Open
Abstract
In the tumor microenvironment, macrophages have been suggested to be stimulated by tumor cells, becoming tumor-associated macrophages that promote cancer development and progression. We examined the effect of these macrophages on human ovarian cancer cell invasion and found that conditioned medium of macrophages stimulated by ovarian cancer cells (OC-MQs) significantly increased cell invasion. CC chemokine ligand 7 (CCL7) expression and production were significantly higher in OC-MQs than in the control macrophages. Peritoneal macrophages from patients with ovarian cancer showed higher CCL7 expression levels than those from healthy controls. Inhibition of CCL7 using siRNA and neutralizing antibodies reduced the OC-MQ-CM-induced ovarian cancer cell invasion. CC chemokine receptor 3 (CCR3) was highly expressed in human ovarian cancer cells, and a specific inhibitor of this receptor reduced the OC-MQ-CM-induced invasion. Specific signaling and transcription factors were associated with enhanced CCL7 expression in OC-MQs. CCL7-induced invasion required the expression of matrix metalloproteinase 9 via activation of extracellular signal-related kinase signaling in human ovarian cancer cells. These data suggest that tumor-associated macrophages can affect human ovarian cancer metastasis via the CCL7/CCR3 axis.
Collapse
|
38
|
Wang SW, Tai HC, Tang CH, Lin LW, Lin TH, Chang AC, Chen PC, Chen YH, Wang PC, Lai YW, Chen SS. Melatonin impedes prostate cancer metastasis by suppressing MMP-13 expression. J Cell Physiol 2021; 236:3979-3990. [PMID: 33251599 DOI: 10.1002/jcp.30150] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/21/2020] [Accepted: 10/24/2020] [Indexed: 12/12/2022]
Abstract
Prostate cancer has high metastatic potential. Men with higher urinary levels of the sleep hormone melatonin are much less likely to develop advanced prostate cancer compared with men with lower levels of melatonin. Melatonin has shown anticancer activity in experimental investigations. Nevertheless, the therapeutic effect of melatonin in metastatic prostate cancer has largely remained a mystery. Analyses of Gene Expression Omnibus data and human tissue samples indicated that levels of matrix metallopeptidase 13 (MMP-13) expression are higher in prostate cancer patients than in healthy cancer-free individuals. Mechanistic investigations revealed that melatonin inhibits MMP-13 expression and the migratory and invasive capacities of prostate cancer cells via the MT1 receptor and the phospholipase C, p38, and c-Jun signaling cascades. Importantly, tumor growth rate and metastasis to distant organs were suppressed by melatonin in an orthotopic prostate cancer model. This is the first demonstration showing that melatonin impedes metastasis of prostate cancer by suppressing MMP-13 expression in both in vitro and in vivo models. Thus, melatonin is promising in the management of prostate cancer metastasis and deserves to undergo clinical investigations.
Collapse
Affiliation(s)
- Shih-Wei Wang
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Huai-Ching Tai
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
- Department of Urology, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Liang-Wei Lin
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Tien-Huang Lin
- Department of Urology, Buddhist Tzu Chi General Hospital Taichung Branch, Taichung, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| | - An-Chen Chang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Po-Chun Chen
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yi-Hsuan Chen
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Department of Urology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Po-Chuan Wang
- Department of Gastroenterology, Hsinchu MacKay Memorial Hospital, Hsinchu City, Taiwan
| | - Yu-Wei Lai
- Division of Urology, Taipei City Hospital Renai Branch, Taipei, Taiwan
- Department of Urology, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Shiou-Sheng Chen
- Department of Urology, National Yang-Ming University School of Medicine, Taipei, Taiwan
- Division of Urology, Taipei City Hospital Zhongxiao Branch, Taipei, Taiwan
- Commission for General Education, College of Applied Science, National Taiwan University of Science and Technology, Taipei, Taiwan
| |
Collapse
|
39
|
Ishikawa M, Iwasaki M, Zhao H, Saito J, Hu C, Sun Q, Sakamoto A, Ma D. Inhalational Anesthetics Inhibit Neuroglioma Cell Proliferation and Migration via miR-138, -210 and -335. Int J Mol Sci 2021; 22:ijms22094355. [PMID: 33919449 PMCID: PMC8122527 DOI: 10.3390/ijms22094355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/17/2021] [Accepted: 04/20/2021] [Indexed: 01/01/2023] Open
Abstract
Inhalational anesthetics was previously reported to suppress glioma cell malignancy but underlying mechanisms remain unclear. The present study aims to investigate the effects of sevoflurane and desflurane on glioma cell malignancy changes via microRNA (miRNA) modulation. The cultured H4 cells were exposed to 3.6% sevoflurane or 10.3% desflurane for 2 h. The miR-138, -210 and -335 expression were determined with qRT-PCR. Cell proliferation and migration were assessed with wound healing assay, Ki67 staining and cell count kit 8 (CCK8) assay with/without miR-138/-210/-335 inhibitor transfections. The miRNA downstream proteins, hypoxia inducible factor-1α (HIF-1α) and matrix metalloproteinase 9 (MMP9), were also determined with immunofluorescent staining. Sevoflurane and desflurane exposure to glioma cells inhibited their proliferation and migration. Sevoflurane exposure increased miR-210 expression whereas desflurane exposure upregulated both miR-138 and miR-335 expressions. The administration of inhibitor of miR-138, -210 or -335 inhibited the suppressing effects of sevoflurane or desflurane on cell proliferation and migration, in line with the HIF-1α and MMP9 expression changes. These data indicated that inhalational anesthetics, sevoflurane and desflurane, inhibited glioma cell malignancy via miRNAs upregulation and their downstream effectors, HIF-1α and MMP9, downregulation. The implication of the current study warrants further study.
Collapse
Affiliation(s)
- Masashi Ishikawa
- Department of Anesthesiology and Pain medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (M.I.); (A.S.)
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
- Correspondence: (M.I.); (D.M.)
| | - Masae Iwasaki
- Department of Anesthesiology and Pain medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (M.I.); (A.S.)
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
| | - Junichi Saito
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
- Department of Anesthesiology, Graduate School of Medicine, Hirosaki University, Hirosaki, Aomori 036-8562, Japan
| | - Cong Hu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
| | - Qizhe Sun
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
| | - Atsuhiro Sakamoto
- Department of Anesthesiology and Pain medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (M.I.); (A.S.)
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
- Correspondence: (M.I.); (D.M.)
| |
Collapse
|
40
|
Jahanbakhshi F, Maleki Dana P, Badehnoosh B, Yousefi B, Mansournia MA, Jahanshahi M, Asemi Z, Halajzadeh J. Curcumin anti-tumor effects on endometrial cancer with focus on its molecular targets. Cancer Cell Int 2021; 21:120. [PMID: 33602218 PMCID: PMC7891161 DOI: 10.1186/s12935-021-01832-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Curcumin is extracted from turmeric and shows a variety of properties that make it a useful agent for treating diseases and targeting different biological mechanisms, including apoptosis, angiogenesis, inflammation, and oxidative stress. This phenolic compound is safe even at high doses. However, it has poor bioavailability. The incidence rates of endometrial cancer (EC) that is one of the most prevalent gynecological malignancies is increasing. Meanwhile, the onset age of EC has been decreased in past few years. Besides, EC does not show a convenient prognosis, particularly at advanced stages. Based on this information, discovering new approaches or enhancing the available ones is required to provide better care for EC patients. In this review, we cover studies concerned with the anti-tumor effects of curcumin on EC. We focus on molecular mechanisms that are targeted by curcumin treatment in different processes of cancer development and progression, such as apoptosis, inflammation, and migration. Furthermore, we present the role of curcumin in targeting some microRNAs (miRNAs) that may play a role in EC.
Collapse
Affiliation(s)
- Fahime Jahanbakhshi
- Department of Gynecology and Obstetrics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Bita Badehnoosh
- Department of Gynecology and Obstetrics, Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Moghadeseh Jahanshahi
- Clinical Research Development Center (CRDC), Sayad Shirazi Hospital, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Jamal Halajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran.
| |
Collapse
|
41
|
Inhibition of platelet-derived growth factor C and their receptors additionally increases doxorubicin effects in triple-negative breast cancer cells. Eur J Pharmacol 2021; 895:173868. [PMID: 33460613 DOI: 10.1016/j.ejphar.2021.173868] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 01/06/2023]
Abstract
Complex of platelet-derived growth factor (PDGF) isoforms and PDGF receptors have important functions in the regulation of growth and survival of various cell types. Herein, it was found that aberrant PDGFC expression is closely associated with survival rates in triple-negative breast cancer (TNBC) patients. In addition, PDGFC expression was identified to be significantly increased in TNBC cells unlike other subtypes such as PDGFA, PDGFB, and PDGFD. Apparently, the effects of specific PDGF receptor (PDGFR) inhibitors such as sunitinib and ponatinib on HCC1806 and Hs578T TNBC cells were investigated. Both inhibitors decreased cell viability in a dose-dependent manner. In addition, the inhibitors completely inhibited cell growth in both the cell lines and decreased the expression of matrix metalloproteinase-1 (MMP-1), one of the metastasis-related genes. Cell migration was also decreased by the inhibitors. Finally, the combined effects of the inhibitors with doxorubicin (DOX) were investigated. The results showed that the combination of two PDGFR inhibitors with DOX inhibited the growth of cells and enhanced the apoptotic cell death more uniformly than DOX. Consequently, it is demonstrated that PDGFR inhibitors, sunitinib and ponatinib hold the potential for effective treatment of TNBC.
Collapse
|
42
|
Park G, Song NY, Kim DH, Lee SJ, Chun KS. Thymoquinone Suppresses Migration of Human Renal Carcinoma Caki-1 Cells through Inhibition of the PGE 2-Mediated Activation of the EP2 Receptor Pathway. Biomol Ther (Seoul) 2021; 29:64-72. [PMID: 32843585 PMCID: PMC7771838 DOI: 10.4062/biomolther.2020.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/07/2020] [Accepted: 07/16/2020] [Indexed: 11/05/2022] Open
Abstract
Renal cell carcinoma (RCC) is likely to metastasize to other organs, and is often resistant to conventional chemotherapies. Thymoquinone (TQ), a phytochemical derived from the seeds of Nigella sativa, has been shown to inhibit migration and metastasis in various cancers. In this study, we assessed the effect of TQ on the migratory activity of human RCC Caki-1 cells. We found that treatment with TQ reduced the proteolytic activity of matrix metalloproteinase-9 (MMP-9) in Caki-1 cells. TQ significantly repressed prostaglandin E2 (PGE2) production, its EP2 receptor expression as well as the activation of Akt and p38, the wellknown upstream signal proteins of MMP-9. In addition, treatment with butaprost, a PGE2 agonist, also induced MMP-9 activity and migration/invasion in Caki-1 cells. Moreover, pharmacological inhibitors of PI3K/Akt and p38 remarkably attenuated butaprostinduced Caki-1 cell migration and invasion, implying that activation of PI3K/Akt and p38 is a bridge between the PGE2-EP2 axis and MMP-9-dependent migration and invasion. Taken together, these data suggest that TQ is a promising anti-metastatic drug to treat advanced and metastatic RCC.
Collapse
Affiliation(s)
- Geumi Park
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Na-Young Song
- College of Dentistry, Younsei University, Seoul 03722, Republic of Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon 16227, Republic of Korea
| | - Su-Jun Lee
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
43
|
Pantazi P, Carollo E, Carter DRF, Brooks SA. A practical toolkit to study aspects of the metastatic cascade in vitro. Acta Histochem 2020; 122:151654. [PMID: 33157489 DOI: 10.1016/j.acthis.2020.151654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/23/2020] [Accepted: 10/28/2020] [Indexed: 12/30/2022]
Abstract
While metastasis - the spread of cancer from the primary location to distant sites in the body - remains the principle cause of cancer death, it is incompletely understood. It is a complex process, requiring the metastatically successful cancer cell to negotiate a formidable series of interconnected steps, which are described in this paper. For each step, we review the range of in vitro assays that may be used to study them. We also provide a range of detailed, step-by-step protocols that can be undertaken in most modestly-equipped laboratories, including methods for converting qualitative observations into quantitative data for analysis. Assays include: (1) a gelatin degradation assay to study the ability of endothelial cells to degrade extracellular matrix during tumour angiogenesis; (2) the morphological characterisation of cells undergoing epithelial-mesenchymal transition (EMT) as they acquire motility; (3) a 'scratch' or 'wound-healing' assay to study cancer cell migration; (4) a transwell assay to study cancer cell invasion through extracellular matrix; and (5) a static adhesion assay to examine cancer cell interactions with, and adhesion to, endothelial monolayers. This toolkit of protocols will enable researchers who are interested in metastasis to begin to focus on defined aspects of the process. It is only by further understanding this complex, fascinating and clinically relevant series of events that we may ultimately devise ways of better treating, or even preventing, cancer metastasis. The assays may also be of more broad interest to researchers interested in studying aspects of cellular behaviour in relation to other developmental and disease processes.
Collapse
|
44
|
Choi C, Jeong W, Ghang B, Park Y, Hyun C, Cho M, Kim J. Cyr61 synthesis is induced by interleukin-6 and promotes migration and invasion of fibroblast-like synoviocytes in rheumatoid arthritis. Arthritis Res Ther 2020; 22:275. [PMID: 33228785 PMCID: PMC7685583 DOI: 10.1186/s13075-020-02369-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
Background Interleukin-6 (IL-6) is involved in fibroblast-like synoviocyte (FLS) activation and promotes pannus formation and bone and cartilage destruction in rheumatoid arthritis (RA). Cysteine-rich 61 (Cyr61) protein regulates cell proliferation, migration, and differentiation. The aim of this study was to investigate the role of Cyr61 in RA-FLS migration and invasion after IL-6 stimulation. Methods Western blotting, immunohistochemistry, reverse transcription-polymerase chain reaction, and real time-polymerase chain reaction were used to examine protein and mRNA levels of Cyr61, matrix metalloproteinases (MMPs), and other signalling proteins. Knockdown of gene expression was performed with siRNA, and RNA sequencing was performed for differential gene analysis. Migration and invasion were assessed by wound healing and Boyden chamber assays. Results Cyr61 levels were elevated in FLSs from RA patients compared to those in osteoarthritis patients. Control and IL-6-treated FLSs showed differential gene expression. IL-6 stimulated protein synthesis of Cyr61, which was attenuated by the extracellular signal-related kinase 1/2 (ERK 1/2) inhibitor, PD98059, and knockdown of early growth response 3 (EGR3), but not of JUN. IL-6-induced Cyr61 protein synthesis increased expression of MMP2. Cyr61 promoted FLS migration and invasion in an autocrine manner. Knockdown of CYR61 and a neutralising antibody attenuated Cyr61 synthesis and IL-6-induced FLS migration. Conclusions By modulating the ERK/EGR3 pathway, IL-6 stimulated Cyr61 production and in turn increased invasiveness of FLS. Our data suggest that Cyr61 might be a potential target to prevent the progression of joint damage in RA.
Collapse
Affiliation(s)
- Changmin Choi
- Department of Medicine, Jeju National University School of Medicine, Jeju, Republic of Korea
| | - Wooseong Jeong
- Department of Internal Medicine, Division of Rheumatology, Jeju National University Hospital, Aran 13gil, Jeju, 690-797, Republic of Korea
| | - Byeongzu Ghang
- Department of Internal Medicine, Division of Rheumatology, Jeju National University Hospital, Aran 13gil, Jeju, 690-797, Republic of Korea
| | - Yonggeun Park
- Department of Orthopaedic Surgery, Jeju National University Hospital, Jeju, Republic of Korea
| | - Changlim Hyun
- Department of Pathology, Jeju National University Hospital, Jeju, Republic of Korea
| | - Moonjae Cho
- Department of Biochemistry, Jeju National University School of Medicine, Aran 13gil, Jeju, 690-797, Republic of Korea.
| | - Jinseok Kim
- Department of Internal Medicine, Division of Rheumatology, Jeju National University Hospital, Aran 13gil, Jeju, 690-797, Republic of Korea.
| |
Collapse
|
45
|
The Novel Target of Colorectal Carcinoma: TRIM44 Regulates Cell Migration and Invasion via Activation of CXCR4/NF-κB Signaling. Cell Biochem Biophys 2020; 79:113-121. [PMID: 33151473 DOI: 10.1007/s12013-020-00955-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 10/23/2022]
Abstract
Tripartite motif containing 44 (TRIM44) has been reported to regulate various biological effects in malignant cancers and matrix Metalloproteinases has been demonstrated to be associated with cancer cell migration and invasion. Nonetheless, the expression and molecular mechanism of TRIM44 in colorectal cancer (CRC) remain rarely known. TRIM44 was overexpressed or knocked down in CRC cells. Subsequently, the effects of TRIM44 on cell migration and invasion as well as underlying molecular mechanisms were detected. Data showed that TRIM44 was highly expressed in CRC cell lines. Downregulation of TRIM44 inhibited the cell viability, migration, and invasion in SW-480 cells. In addition, overexpression of TRIM44 enhanced the expression of NF-κB and CXCR4, and enhanced the binding between NF-κB and CXCR4 promoter region. In summarize, TRIM44 may serve as a potential target for CRC diagnosis and progression.
Collapse
|
46
|
Xiaoxia X, Jing S, Dongbin X, Yonggang T, Jingke Z, Yanying Z, Hulai W. Realgar Nanoparticles Inhibit Migration, Invasion and Metastasis in a Mouse Model of Breast Cancer by Suppressing Matrix Metalloproteinases and Angiogenesis. Curr Drug Deliv 2020; 17:148-158. [PMID: 31939730 DOI: 10.2174/1567201817666200115105633] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/21/2019] [Accepted: 12/31/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Realgar, a traditional Chinese medicine, has shown antitumor efficacy in several tumor types. We previously showed that realgar nanoparticles (nano-realgar) had significant antileukemia, anti-lung cancer and anti-liver cancer effects. In addition, the anti-tumor effects of nanorealgar were significantly better than those of ordinary realgar. OBJECTIVE To explore the inhibitory effects and molecular mechanisms of nano-realgar on the migration, invasion and metastasis of mouse breast cancer cells. METHODS Wound-healing migration assays and Transwell invasion assays were carried out to determine the effects of nano-realgar on breast cancer cell (4T1) migration and invasion. The expression levels of matrix metalloproteinase (MMP)-2 and -9 were measured by Western blot. A murine breast cancer metastasis model was established, administered nano-realgar for 32 days and monitored for tumor growth and metastasis by an in vivo optical imaging system. Finally, living imaging and hematoxylin and eosin (HE) staining were used to measure the morphology and pathology of lung and liver cancer cell metastases, respectively. Angiogenesis was assessed by CD34 immunohistochemistry. RESULTS Nano-realgar significantly inhibited the migration and invasion of breast cancer 4T1 cells and the expression of MMP-2 and -9. Meanwhile, nano-realgar effectively suppressed the abilities of tumor growth, metastasis and angiogenesis in the murine breast cancer metastasis model in a time- and dosedependent manner. CONCLUSION Nano-realgar significantly inhibited migration and invasion of mouse breast cancer cells in vitro as well as pulmonary and hepatic metastasis in vivo, which may be closely correlated with the downexpression of MMP-2 and -9 and suppression of tumor neovascularization.
Collapse
Affiliation(s)
- Xi Xiaoxia
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Sun Jing
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xi Dongbin
- General Surgery Department, People's Hospital of Jiuquan City, Jiuquan, China
| | - Tian Yonggang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Zhang Jingke
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Zhang Yanying
- Laboratory Animal Center, Gansu University of Chinese Medicine, Lanzhou, China
| | - Wei Hulai
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
47
|
Liu R, Li H, Xu Y, Li X, Guo X, Shi J, Cui Y, Wang Z, Liu J. Blockade of TRIM59 enhances esophageal cancer cell chemosensitivity to cisplatin by upregulating p53. Oncol Lett 2020; 21:6. [PMID: 33240412 PMCID: PMC7681221 DOI: 10.3892/ol.2020.12267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
Human esophageal cancer (hESC) cell motility adopts various modes, resulting in hESC progression and poor survival. However, how tripartite motif 59 (TRIM59), as the ubiquitination machinery, participates in hESC metastasis is not completely understood. The results indicated that TRIM59 was aberrantly upregulated in hESC tissues compared with adjacent healthy esophageal tissues, which was associated with poor survival and advanced TNM state among patients with hESC. Moreover, patients with hESC with higher TRIM59 expression displayed undetectable p53 expression, which contributed to enhanced progression and motility of hESC. At the molecular level, TRIM59 was indicated to be an E3 putative ubiquitin ligase that targeted the p53 protein, leading to increased degradation of p53, which resulted in decreased chemosensitivity to cisplatin. TRIM59 knockdown reduced TRIM59 expression, increased p53 protein expression, and decreased hESC cell viability, clone formation and migration compared with the small interfering RNA negative control (siNC) group. Furthermore, hESC cell lines were more sensitive to cisplatin in the TRIM59-knockdown group compared with the siNC group. The results indicated a relationship between TRIM59, p53 and the chemosensitivity of cisplatin. The present study suggested that TRIM59 may serve as a promising prognostic indicator for patients with hESC.
Collapse
Affiliation(s)
- Rongfeng Liu
- Department of Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Hongchen Li
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Yanzhao Xu
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xing Li
- Department of Immuno-Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaojin Guo
- Department of Immuno-Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jian Shi
- Department of Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yanzhi Cui
- Department of Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Zhiyu Wang
- Department of Immuno-Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Junfeng Liu
- Third Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
48
|
Wei X, Yang W, Zhang F, Cheng F, Rao J, Lu L. PIGU promotes hepatocellular carcinoma progression through activating NF-κB pathway and increasing immune escape. Life Sci 2020; 260:118476. [PMID: 32971102 DOI: 10.1016/j.lfs.2020.118476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignancy and has the third highest mortality rate among all tumors. Previous studies found that phosphatidylinositol glycan anchor biosynthesis class U (PIGU) was highly expressed in hepatocellular carcinoma (HCC), while the function of PIGU in HCC remains unknown. Here, we deeply investigated this issue. The expression levels of PIGU in HCC cells were measured by Western blotting. The functions of PIGU in HCC cells were assessed in vitro, followed by assessing the nuclear factor-kappa B (NF-κB) pathway-related protein levels. The xenograft mouse models were conducted to investigate the effects of PIGU in vivo. Moreover, the effects of PIGU downregulation on natural killer (NK)-92 cell-mediated cell killing were detected. The results showed that PIGU was highly expressed in HCC cells compared with normal liver cells. Functional studies showed that PIGU promoted viability, cell cycle progression, migration, and invasion and suppressed apoptosis in HCC cells. Mechanism studies indicated that PIGU silencing blocked the NF-κB pathway and the blockade of the NF-κB pathway reversed the effects of PIGU overexpression on HCC cell function, including cell viability, migration, invasion, and apoptosis. In vivo studies further verified the effects of PIGU on HCC cell function, and demonstrated that PIGU knockdown suppressed tumorigenesis. Additionally, we proved that PIGU downregulation significantly enhanced the sensitivity of HCC cells to NK-92 cell cytolysis. Collectively, PIGU may promote HCC progression through activating the NF-κB pathway and promoting immune escape, indicating that PIGU may serve as a promising therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Xin Wei
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing 210029, China
| | - Wenjie Yang
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing 210029, China
| | - Feng Zhang
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing 210029, China
| | - Feng Cheng
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing 210029, China
| | - Jianhua Rao
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing 210029, China.
| | - Ling Lu
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing 210029, China.
| |
Collapse
|
49
|
Kansakar U, Wang W, Markovic V, Sossey-Alaoui K. Elucidating the molecular signaling pathways of WAVE3. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:900. [PMID: 32793744 DOI: 10.21037/atm.2020.02.16] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer metastasis is a complex, multistep process that requires tumor cells to evade from the original site and form new tumors at a distant site or a different organ, often via bloodstream or the lymphatic system. Metastasis is responsible for more than 90% of cancer-related deaths. WAVE3 belongs to the Wiskott-Aldrich syndrome protein (WASP) family, which regulate actin cytoskeleton remodeling as well as several aspects of cell migration, invasion, and metastasis. In fact, WAVE3 has been established as a driver of tumor progression and metastasis in cancers from several origins, including triple negative breast cancers (TNBCs), which are classified as the most lethal subtype of breast cancer, due to their resistance to standard of care therapy and highly metastatic behavior. In this review, we will attempt to summarize the recent advances that have been made to understand how WAVE3 contributes to the molecular mechanisms that control cancer progression and metastasis. We will also review the signaling pathways that are involved in the regulation of WAVE3 expression and function to identify potential therapeutic options targeted against WAVE3 for the treatment of patients with metastatic tumors.
Collapse
Affiliation(s)
- Urna Kansakar
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rammelkamp Center for Research, MetroHealth, Cleveland, OH, USA
| | - Wei Wang
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rammelkamp Center for Research, MetroHealth, Cleveland, OH, USA
| | - Vesna Markovic
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rammelkamp Center for Research, MetroHealth, Cleveland, OH, USA
| | - Khalid Sossey-Alaoui
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rammelkamp Center for Research, MetroHealth, Cleveland, OH, USA
| |
Collapse
|
50
|
Avagliano A, Fiume G, Ruocco MR, Martucci N, Vecchio E, Insabato L, Russo D, Accurso A, Masone S, Montagnani S, Arcucci A. Influence of Fibroblasts on Mammary Gland Development, Breast Cancer Microenvironment Remodeling, and Cancer Cell Dissemination. Cancers (Basel) 2020; 12:E1697. [PMID: 32604738 PMCID: PMC7352995 DOI: 10.3390/cancers12061697] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
The stromal microenvironment regulates mammary gland development and tumorigenesis. In normal mammary glands, the stromal microenvironment encompasses the ducts and contains fibroblasts, the main regulators of branching morphogenesis. Understanding the way fibroblast signaling pathways regulate mammary gland development may offer insights into the mechanisms of breast cancer (BC) biology. In fact, the unregulated mammary fibroblast signaling pathways, associated with alterations in extracellular matrix (ECM) remodeling and branching morphogenesis, drive breast cancer microenvironment (BCM) remodeling and cancer growth. The BCM comprises a very heterogeneous tissue containing non-cancer stromal cells, namely, breast cancer-associated fibroblasts (BCAFs), which represent most of the tumor mass. Moreover, the different components of the BCM highly interact with cancer cells, thereby generating a tightly intertwined network. In particular, BC cells activate recruited normal fibroblasts in BCAFs, which, in turn, promote BCM remodeling and metastasis. Thus, comparing the roles of normal fibroblasts and BCAFs in the physiological and metastatic processes, could provide a deeper understanding of the signaling pathways regulating BC dissemination. Here, we review the latest literature describing the structure of the mammary gland and the BCM and summarize the influence of epithelial-mesenchymal transition (EpMT) and autophagy in BC dissemination. Finally, we discuss the roles of fibroblasts and BCAFs in mammary gland development and BCM remodeling, respectively.
Collapse
Affiliation(s)
- Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy;
| | - Nunzia Martucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Eleonora Vecchio
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Luigi Insabato
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Daniela Russo
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Antonello Accurso
- Department of General, Oncological, Bariatric and Endocrine-Metabolic Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Montagnani
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| |
Collapse
|