151
|
Sayer S, Zandrini T, Markovic M, Van Hoorick J, Van Vlierberghe S, Baudis S, Holnthoner W, Ovsianikov A. Guiding cell migration in 3D with high-resolution photografting. Sci Rep 2022; 12:8626. [PMID: 35606455 PMCID: PMC9126875 DOI: 10.1038/s41598-022-11612-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/24/2022] [Indexed: 11/09/2022] Open
Abstract
Multi-photon lithography (MPL) has proven to be a suitable tool to precisely control the microenvironment of cells in terms of the biochemical and biophysical properties of the hydrogel matrix. In this work, we present a novel method, based on multi-photon photografting of 4,4′-diazido-2,2′-stilbenedisulfonic acid (DSSA), and its capabilities to induce cell alignment, directional cell migration and endothelial sprouting in a gelatin-based hydrogel matrix. DSSA-photografting allows for the fabrication of complex patterns at a high-resolution and is a biocompatible, universally applicable and straightforward process that is comparably fast. We have demonstrated the preferential orientation of human adipose-derived stem cells (hASCs) in response to a photografted pattern. Co-culture spheroids of hASCs and human umbilical vein endothelial cells (HUVECs) have been utilized to study the directional migration of hASCs into the modified regions. Subsequently, we have highlighted the dependence of endothelial sprouting on the presence of hASCs and demonstrated the potential of photografting to control the direction of the sprouts. MPL-induced DSSA-photografting has been established as a promising method to selectively alter the microenvironment of cells.
Collapse
Affiliation(s)
- Simon Sayer
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria.,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria
| | - Tommaso Zandrini
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria.,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria
| | - Marica Markovic
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria.,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria
| | - Jasper Van Hoorick
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Stefan Baudis
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria.,Polymer Chemistry and Technology Group, Institute of Applied Synthetic Chemistry, TU Wien, Vienna, Austria
| | - Wolfgang Holnthoner
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria.,Ludwig-Boltzmann-Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
| | - Aleksandr Ovsianikov
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria. .,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria.
| |
Collapse
|
152
|
Cevik M, Namal E, Dinc-Sener N, Iner-Koksal U, Ciftci C, Susleyici B. Investigation of Vascular Endothelial Growth Factor Polymorphisms on Risk, Metastasis, Laterality, and Prognosis of Colorectal Cancer in Turkish Subjects. Genet Test Mol Biomarkers 2022; 26:298-306. [PMID: 35593899 DOI: 10.1089/gtmb.2021.0213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objectives: Tumor angiogenesis is known to support the spread and invasion of tumor cells, allow distant organ metastasis, resulting in worse prognosis and mortality. Since vascular endothelial growth factor-A (VEGF-A) is the major regulator of angiogenesis, in the present study, the associations of VEGF-A +405G>C and -460C>T polymorphisms with risk, primary tumor location, prognosis, and metastasis of colorectal cancer (CRC) were investigated in Turkish subjects. Material and Methods: A total of 153 subjects consisting of 74 controls and 79 CRC diagnosed patients were included in the study. VEGF-A +405G>C and -460C>T polymorphisms were analyzed using Agena MassARRAY platform. Results: VEGF +405GC+CC genotypes were found to be significantly associated with left colon cancer (unadjusted odds ratio [OR] = 5.208 confidence interval [95% CI]: 1.064-25.496, p = 0.04). VEGF -460TT and CT+TT genotypes were associated with reduced liver metastasis risk (OR = 0.080 95% CI: 0.009-0.689 p = 0.02 and OR = 0.191 95% CI: 0.039-0.925, p = 0.04, respectively). Patients with VEGF +405GG genotype showed longer progression-free survival as a response to bevacizumab treatment (Log rank = 6.92, p = 0.03). Conclusion: According to our results, VEGF +405G>C and -460C>T polymorphisms were found to be associated with CRC prognosis, sidedness, and metastasis. Our findings should be conducted in further studies.
Collapse
Affiliation(s)
- Mehtap Cevik
- Department of Molecular Biology, Marmara University Faculty of Science and Letters, Istanbul, Turkey
| | - Esat Namal
- Department of Medical Oncology, Demiroglu Bilim University Faculty of Medicine, Istanbul, Turkey
| | - Nur Dinc-Sener
- Department of Medical Oncology, Demiroglu Bilim University Faculty of Medicine, Istanbul, Turkey
| | | | - Cavlan Ciftci
- Department of Cardiology, Demiroglu Bilim University Faculty of Medicine, Istanbul, Turkey
| | - Belgin Susleyici
- Department of Molecular Biology, Marmara University Faculty of Science and Letters, Istanbul, Turkey
| |
Collapse
|
153
|
Shinde VR, Revi N, Murugappan S, Singh SP, Rengan AK. Enhanced Permeability and Retention Effect: A key facilitator for solid tumor targeting by nanoparticles. Photodiagnosis Photodyn Ther 2022; 39:102915. [PMID: 35597441 DOI: 10.1016/j.pdpdt.2022.102915] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 12/14/2022]
Abstract
Exploring the enhanced permeability and retention (EPR) effect through therapeutic nanoparticles has been a subject of considerable interest in tumor biology. This passive targeting based phenomenon exploits the leaky blood vasculature and the defective lymphatic drainage system of the heterogeneous tumor microenvironment resulting in enhanced preferential accumulation of the nanoparticles within the tumor tissues. This article reviews the fundamental studies to assess how the EPR effect plays an essential role in passive targeting. Further, it summarizes various therapeutic modalities of nanoformulation including chemo-photodynamic therapy, intravascular drug release, and photothermal immunotherapy to combat cancer using enhanced EPR effect in neoplasia region.
Collapse
Affiliation(s)
- Vinod Ravasaheb Shinde
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Neeraja Revi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | | | - Surya Prakash Singh
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India.
| |
Collapse
|
154
|
Wu Y, Xu X, Liu M, Qin X, Wu Q, Ding H, Zhao Q. DZW-310, a novel phosphoinositide 3-kinase inhibitor, attenuates the angiogenesis and growth of hepatocellular carcinoma cells via PI3K/AKT/mTOR axis. Biochem Pharmacol 2022; 201:115093. [PMID: 35580648 DOI: 10.1016/j.bcp.2022.115093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/02/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most malignant tumors worldwide with high lethality and prevalence. The deregulated phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway plays an indispensable role in mediating the progression of HCC. Among them, PI3K acts as the most pivotal initiator, contributing to multiple malignant biological processes, like proliferation, apoptosis and angiogenesis. Many PI3K inhibitors (PI3Kis) have been proved or proceeded into clinical as antineoplastic drugs. Nevertheless, the application of PI3Kis for the treatment of HCC remains a blank. Accordingly, our study identified a novel PI3Ki (DZW-310) with strong anti-HCC activity in vitro and in vivo. This study aimed to evaluate its anti-HCC effect and elucidate its potential mechanism. Our current results revealed that DZW-310 significantly attenuated HCC cell growth through promoting intrinsic apoptosis and G0/G1 phase cell arrest. Moreover, DZW-310 suppressed angiogenesis by regulating the HIF-1α/VEGFA axis. Further mechanistic investigation demonstrated that DZW-310, functioned as a PI3Ki, exerted strong anti-HCC activity by acting on PI3Kα (a major subtype of PI3K) and ulteriorly deactivating the PI3K/AKT/mTOR pathway. Collectively, our studies identified that DZW-310 is expected to become a promising HCC therapeutic agent and broaden clinical application of PI3Ki in HCC chemotherapy.
Collapse
Affiliation(s)
- Yuting Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, 110016, PR China.
| | - Xiangbo Xu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, 110016, PR China.
| | - Mingyue Liu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, 110016, PR China.
| | - Xiaochun Qin
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, 110016, PR China.
| | - Qiong Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, 110016, PR China.
| | - Huaiwei Ding
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, 110016, PR China; State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong Province, 518055, PR China.
| | - Qingchun Zhao
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, 110016, PR China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110840, PR China.
| |
Collapse
|
155
|
Ravi S, Alencar AM, Arakelyan J, Xu W, Stauber R, Wang CCI, Papyan R, Ghazaryan N, Pereira RM. An Update to Hallmarks of Cancer. Cureus 2022; 14:e24803. [PMID: 35686268 PMCID: PMC9169686 DOI: 10.7759/cureus.24803] [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] [Accepted: 05/06/2022] [Indexed: 12/03/2022] Open
Abstract
In the last decade, there has been remarkable progress in research toward understanding and refining the hallmarks of cancer. In this review, we propose a new hallmark - "pro-survival autophagy." The importance of pro-survival autophagy is well established in tumorigenesis, as it is related to multiple steps in cancer progression and vital for some cancers. Autophagy is a potential anti-cancer therapeutic target. For this reason, autophagy is a good candidate as a new hallmark of cancer. We describe two enabling characteristics that play a major role in enabling cells to acquire the hallmarks of cancer - "tumor-promoting microenvironment and macroenvironment" and "cancer epigenetics, genome instability and mutation." We also discuss the recent updates, therapeutic and prognostic implications of the eight hallmarks of cancer described by Hanahan et al. in 2011. Understanding these hallmarks and enabling characteristics is key not only to developing new ways to treat cancer efficiently but also to exploring options to overcome cancer resistance to treatment.
Collapse
Affiliation(s)
- Swapna Ravi
- Department of Medicine, St. Luke's Hospital, Duluth, USA
| | - Antonio M Alencar
- Department of Medical Oncology, Hospital Universitário da Universidade Federal do Maranhão, Hospital São Domingos, São Luís, BRA
| | - Jemma Arakelyan
- Department of Oncology/Solid Tumors, Yerevan State Medical University, Hematology Center After Prof. R. Yeolyan, Yerevan, ARM
| | - Weihao Xu
- Department of Business Development, Harbour BioMed, Boston, USA
| | - Roberta Stauber
- Department of Oncology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, BRA
| | - Cheng-Chi I Wang
- Department of Research and Development, Beltie Bio, Inc, San Diego, USA
| | - Ruzanna Papyan
- Department of Pediatric Oncology and Hematology, Yerevan State Medical University, Pediatric Center and Blood Disorders Center of Armenia, Yerevan, ARM
| | - Narine Ghazaryan
- Department of Molecular Biology, L.A. Orbeli Institute of Physiology National Academy of Sciences, Republic of Armenia (NAS RA) Hematology Center After Prof. R. Yeolyan, Yerevan, ARM
| | - Rosalina M Pereira
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, USA
| |
Collapse
|
156
|
Inositol hexakisphosphate induces apoptosis, cell cycle arrest in non-Hodgkin’s Burkitt lymphoma cells and mediates anti-angiogenic, antitumor effects in T-cell lymphoma bearing Swiss albino mice. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
157
|
Singh V, Khan N, Jayandharan GR. Vector engineering, strategies and targets in cancer gene therapy. Cancer Gene Ther 2022; 29:402-417. [PMID: 33859378 DOI: 10.1038/s41417-021-00331-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 02/23/2021] [Accepted: 03/24/2021] [Indexed: 02/02/2023]
Abstract
Understanding the molecular basis of disease and the design of rationally designed molecular therapies has been the holy grail in the management of human cancers. Gene-based therapies are an important avenue for achieving a possible cure. Focused research in the last three decades has provided significant clues to optimize the potential of cancer gene therapy. The development of gene therapies with a high potential to kill the target cells at the lowest effective dose possible, the development of vectors with significant ability to target cancer-associated antigen, the application of adjunct therapies to target dysregulated microRNA, and embracing a hybrid strategy with a combination of gene therapy and low-dose chemotherapy in a disease-specific manner will be pivotal. This article outlines the advances and challenges in the field with emphasis on the biology and scope of vectors used for gene transfer, newer targets identified, and their outcome in preclinical and clinical studies.
Collapse
Affiliation(s)
- Vijayata Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, UP, India
| | - Nusrat Khan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, UP, India
| | - Giridhara R Jayandharan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, UP, India. .,The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology, Kanpur, UP, India.
| |
Collapse
|
158
|
Yin J, Yang G, Qin X, Li H, Wang L. Optimized U-Net model for 3D light-sheet image segmentation of zebrafish trunk vessels. BIOMEDICAL OPTICS EXPRESS 2022; 13:2896-2908. [PMID: 35774342 PMCID: PMC9203114 DOI: 10.1364/boe.449714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/11/2022] [Accepted: 03/29/2022] [Indexed: 06/15/2023]
Abstract
The growth of zebrafish's vessels can be used as an indicator of the vascular development process and to study the biological mechanisms. The three-dimensional (3D) structures of zebrafish's trunk vessels could be imaged by state-of-art light-sheet fluorescent microscopy with high efficiency. A large amount of data was then produced. Accurate segmentation of these 3D images becomes a new bottleneck for automatic and quantitative analysis. Here, we propose a Multi-scale 3D U-Net model to perform the segmentation of trunk vessels. The segmentation accuracies of 82.3% and 83.0%, as evaluated by the IoU (Intersection over Union) parameter, were achieved for intersegmental vessels and the dorsal longitudinal anastomotic vessels respectively. The growth of zebrafish vasculature from 42-62 hours was then analyzed quantitatively.
Collapse
Affiliation(s)
- Jingyi Yin
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Jiangsu Key Laboratory of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China
| | - Guang Yang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xiaofei Qin
- Jiangsu Key Laboratory of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China
| | - Hui Li
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Jiangsu Key Laboratory of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China
| | - Linbo Wang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
159
|
Tumor microenvironment as a metapopulation model: the effects of angiogenesis, emigration and treatment modalities. J Theor Biol 2022; 545:111147. [PMID: 35489642 DOI: 10.1016/j.jtbi.2022.111147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 03/21/2022] [Accepted: 04/21/2022] [Indexed: 11/21/2022]
Abstract
Tumors consist of heterogeneous cell subpopulations that may develop differing phenotypes, such as increased cell growth, metastatic potential and treatment sensitivity or resistance. To study the dynamics of cancer development at a single-cell level, we model the tumor microenvironment as a metapopulation, in which habitat patches correspond to possible sites for cell subpopulations. Cancer cells may emigrate into dispersal pool (e.g. circulation system) and spread to new sites (i.e. metastatic disease). In the patches, cells divide and new variants may arise, possibly leading into an invasion provided the aberration promotes the cell growth. To study such adaptive landscape of cancer ecosystem, we consider various evolutionary strategies (phenotypes), such as emigration and angiogenesis, which are important determinants during early stages of tumor development. We use the metapopulation fitness of new variants to investigate how these strategies evolve through natural selection and disease progression. We further study various treatment effects and investigate how different therapy regimens affect the evolution of the cell populations. These aspects are relevant, for example, when examining the dynamic process of a benign tumor becoming cancerous, and what is the best treatment strategy during the early stages of cancer development. It is shown that positive angiogenesis promotes cancer cell growth in the absence of anti-angiogenic treatment, and that the anti-angiogenic treatment reduces the need of cytotoxic treatment when used in a combination. Interestingly, the model predicts that treatment resistance might become a favorable quality to cancer cells when the anti-angiogenic treatment is intensive enough. Thus, the optimal treatment dosage should remain below a patient-specific level to avoid treatment resistance.
Collapse
|
160
|
Yao L, Li Y, Chen H, Wen X, Pang Y, Chen Z, Guo Z, Zhang X, Wu H, Guo W. Dual Targeting of Integrin α vβ 3 and Neuropilin-1 Receptors Improves Micropositron Emission Tomography Imaging of Breast Cancer. Mol Pharm 2022; 19:1458-1467. [PMID: 35414180 DOI: 10.1021/acs.molpharmaceut.1c01015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The receptors neuropilin-1 (NRP-1) and integrin αvβ3 are overexpressed in breast cancer and associated with neovascularization. We synthesized a heterodimeric tracer, 68Ga-DOTA-RGD-ATWLPPR, which simultaneously targets integrin αvβ3 and NRP-1 in breast cancer. In this study, we evaluated the diagnostic efficacy of 68Ga-DOTA-RGD-ATWLPPR during micropositron emission tomography (microPET)/X-ray computed tomography (CT) imaging and gamma counting. We evaluated the receptor-binding characteristics and tumor-targeting efficacy of the tracer in vitro and in vivo. Static microPET/CT imaging and gamma counting studies showed that 68Ga-DOTA-RGD-ATWLPPR uptake in MCF-7 tumors is higher than that of monomeric tracers. 68Ga-DOTA-RGD-ATWLPPR uptake could be blocked with excess unlabeled RGD or ATWLPPR, demonstrating the sensitivity and specificity of the tracer. We did not observe bone tracer uptake in vivo, but the data indicated that 68Ga-DOTA-RGD-ATWLPPR is metabolized in the kidneys and the liver uptake is low. In conclusion, 68Ga-DOTA-RGD-ATWLPPR has improved binding affinity, targeting efficiency, and tumor retention time when compared to monomeric tracers, suggesting that it has potential as an imaging probe for breast cancer detection.
Collapse
Affiliation(s)
- Lanlin Yao
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Haojun Chen
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Xuejun Wen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yizhen Pang
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zijun Chen
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Hua Wu
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Wei Guo
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| |
Collapse
|
161
|
Endothelial Cell Metabolism in Vascular Functions. Cancers (Basel) 2022; 14:cancers14081929. [PMID: 35454836 PMCID: PMC9031281 DOI: 10.3390/cancers14081929] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Recent findings in the field of vascular biology are nourishing the idea that targeting the endothelial cell metabolism may be an alternative strategy to antiangiogenic therapy, as well as a novel therapeutic approach for cardiovascular disease. Deepening the molecular mechanisms regulating how ECs re-adapt their metabolic status in response to the changeable conditions of the tissue microenvironment may be beneficial to develop novel innovative treatments to counteract the aberrant growth of vasculature. Abstract The endothelium is the innermost layer of all blood and lymphatic vessels composed of a monolayer of specialized endothelial cells (ECs). It is regarded as a dynamic and multifunctional endocrine organ that takes part in essential processes, such as the control of blood fluidity, the modulation of vascular tone, the regulation of immune response and leukocyte trafficking into perivascular tissues, and angiogenesis. The inability of ECs to perform their normal biological functions, known as endothelial dysfunction, is multi-factorial; for instance, it implicates the failure of ECs to support the normal antithrombotic and anti-inflammatory status, resulting in the onset of unfavorable cardiovascular conditions such as atherosclerosis, coronary artery disease, hypertension, heart problems, and other vascular pathologies. Notably, it is emerging that the ability of ECs to adapt their metabolic status to persistent changes of the tissue microenvironment could be vital for the maintenance of vascular functions and to prevent adverse vascular events. The main purpose of the present article is to shed light on the unique metabolic plasticity of ECs as a prospective therapeutic target; this may lead to the development of novel strategies for cardiovascular diseases and cancer.
Collapse
|
162
|
Loggenberg SR, Twilley D, De Canha MN, Meyer D, Mabena EC, Lall N. Evaluation of Wound Healing and Antibacterial Potential of Greyia radlkoferi Szyszyl. Ethanolic Leaf Extract. Front Pharmacol 2022; 13:806285. [PMID: 35479311 PMCID: PMC9035865 DOI: 10.3389/fphar.2022.806285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/02/2022] [Indexed: 11/16/2022] Open
Abstract
Angiogenesis is an essential mechanism in both physiological and pathological functions, such as wound healing and cancer metastasis. Several growth factors mediate angiogenesis, including vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF). This study evaluated the potential wound healing activity of Greyia radlkoferi Szyszyl (GR) and its effect on growth factors regulating angiogenesis. The ethanolic leaf extract of GR was evaluated for antibacterial activity against wound associated bacteria; Staphylococcus aureus and Pseudomonas aeruginosa. It exhibited antibacterial activity against two strains of S. aureus (ATCC 25293 and ATCC 6538) displaying a minimum inhibitory concentration (MIC) at 250 and 500 μg/ml, respectively. The antioxidant activity of the extract was investigated for nitric oxide (NO) scavenging activity and showed a fifty percent inhibitory concentration (IC50) of 1266.5 ± 243.95 μg/ml. The extract was further investigated to determine its effect on the proliferation and modulation of growth factors secreted by human keratinocytes (HaCaT). Its effect on wound closure was evaluated using the scratch assay, where non-toxic concentrations were tested, as determined by the antiproliferative assay against HaCat cells (IC50 > 400 μg/ml). Results showed that the extract significantly inhibited wound closure, with a percentage closure of 60.15 ± 1.41% (p < 0.05) and 49.52 ± 1.43% (p < 0.01) at a concentration of 50 and 100 μg/ml, respectively, when compared to the 0.25% Dimethyl sulfoxide vehicle control (65.86 ± 1.12%). Quantification of secreted growth factors from cell-free supernatant, collected from the scratch assay, revealed that the extract significantly decreased the concentration of platelet-derived growth factor (PDGF-AA) at both 50 (p < 0.05) and 100 μg/ml (p < 0.001) (443.08 ± 77.36 and 178.98 ± 36.60 pg/ml) when compared to the 0.25% DMSO vehicle control (538.33 ± 12.64 pg/ml). Therefore, whilst the extract showed antibacterial activity against wound associated bacteria, it did not induce wound healing but rather showed a significant inhibition of wound closure, which was confirmed by the inhibition of PDGF-AA, a major growth factor involved in angiogenesis. Therefore, the GR extract, should be considered for further investigation of anti-angiogenic and anti-metastatic properties against cancer cells.
Collapse
Affiliation(s)
| | - Danielle Twilley
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria, South Africa
| | - Marco Nuno De Canha
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria, South Africa
| | - Debra Meyer
- Department of Biochemistry, Faculty of Science, University of Johannesburg, Johannesburg, South Africa
| | | | - Namrita Lall
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria, South Africa
- School of Natural Resources, University of Missouri, Columbia, MO, United States
- College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
- Bio-Tech R and D Institute, University of the West Indies, Kingston, Jamaica
- *Correspondence: Namrita Lall,
| |
Collapse
|
163
|
Escobar-Puentes AA, Palomo I, Rodríguez L, Fuentes E, Villegas-Ochoa MA, González-Aguilar GA, Olivas-Aguirre FJ, Wall-Medrano A. Sweet Potato (Ipomoea batatas L.) Phenotypes: From Agroindustry to Health Effects. Foods 2022; 11:foods11071058. [PMID: 35407143 PMCID: PMC8997864 DOI: 10.3390/foods11071058] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/02/2022] [Accepted: 04/03/2022] [Indexed: 02/04/2023] Open
Abstract
Sweet potato (SP; Ipomoea batatas (L.) Lam) is an edible tuber native to America and the sixth most important food crop worldwide. China leads its production in a global market of USD 45 trillion. SP domesticated varieties differ in specific phenotypic/genotypic traits, yet all of them are rich in sugars, slow digestible/resistant starch, vitamins, minerals, bioactive proteins and lipids, carotenoids, polyphenols, ascorbic acid, alkaloids, coumarins, and saponins, in a genotype-dependent manner. Individually or synergistically, SP’s phytochemicals help to prevent many illnesses, including certain types of cancers and cardiovascular disorders. These and other topics, including the production and market diversification of raw SP and its products, and SP’s starch as a functional ingredient, are briefly discussed in this review.
Collapse
Affiliation(s)
- Alberto A. Escobar-Puentes
- Biomedical Sciences Institute, Autonomous University of Ciudad Juárez, Anillo envolvente del Pronaf y Estocolmo s/n, Ciudad Juárez 32300, Chihuahua, Mexico; or
- Faculty of Medicine and Psychology, Autonomous University of Baja California, Tijuana 22427, Baja California, Mexico
| | - Iván Palomo
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, University of Talca, Talca 3460000, Chile; (I.P.); (L.R.); (E.F.)
| | - Lyanne Rodríguez
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, University of Talca, Talca 3460000, Chile; (I.P.); (L.R.); (E.F.)
| | - Eduardo Fuentes
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, University of Talca, Talca 3460000, Chile; (I.P.); (L.R.); (E.F.)
| | - Mónica A. Villegas-Ochoa
- Center for Research on Food and Development, Carretera al ejido la Victoria Km 0.6, Hermosillo 83304, Sonora, Mexico; (M.A.V.-O.); (G.A.G.-A.)
| | - Gustavo A. González-Aguilar
- Center for Research on Food and Development, Carretera al ejido la Victoria Km 0.6, Hermosillo 83304, Sonora, Mexico; (M.A.V.-O.); (G.A.G.-A.)
| | - Francisco J. Olivas-Aguirre
- Department of Health Sciences, University of Sonora (Campus Cajeme), Blvd. Bordo Nuevo s/n, 7 Ejido Providencia, Cd. Obregón 85199, Sonora, Mexico
- Correspondence: (F.J.O.-A.); (A.W.-M.); Tel.: +52-(656)-3058685 (F.J.O.-A.); +52-(656)-3052344 (A.W.-M.)
| | - Abraham Wall-Medrano
- Biomedical Sciences Institute, Autonomous University of Ciudad Juárez, Anillo envolvente del Pronaf y Estocolmo s/n, Ciudad Juárez 32300, Chihuahua, Mexico; or
- Correspondence: (F.J.O.-A.); (A.W.-M.); Tel.: +52-(656)-3058685 (F.J.O.-A.); +52-(656)-3052344 (A.W.-M.)
| |
Collapse
|
164
|
Hu Z, Sun S, Zhao X, Yu H, Wu X, Wang J, Chang J, Wang H. Rh-Endostatin Plus Irinotecan/Cisplatin as Second-Line Therapy for Advanced Esophageal Squamous Cell Carcinoma: An Open-Label, Phase II Study. Oncologist 2022; 27:253-e312. [PMID: 35380726 PMCID: PMC8982371 DOI: 10.1093/oncolo/oyab078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND This open-label, phase II study aimed to investigate the efficacy and safety of recombinant human endostatin (Rh-endostatin) plus irinotecan/cisplatin as second-line treatment in patients with advanced esophageal squamous cell carcinoma (ESCC). METHODS Eligible patients received 15mg/m2 Rh-endostatin as a continuous intravenous pump infusion (7 continuous days), 60mg/m2 irinotecan (days 1 and 8), and 60mg/m2 cisplatin (day 1) every 3 weeks. The primary endpoint was progression-free survival (PFS). RESULTS A total of 50 patients were assessable for efficacy and safety analysis. The median follow-up was 10.97 months (95%CI: 7.03-19.42) as the data cutoff. Median PFS was 4.01 months (95% CI: 3.19-5.49), and median overall survival (OS) was 12.32 months (95% CI: 8.21-17.45); 13 (26%; 95% CI: 15.87-39.55) of 50 patients had an objective response, and 31 (62%; 95% CI: 48.15-74.14) had disease control. Grade 3 or greater treatment-related adverse events (AEs) occurred in 12 (24.0%) patients, and no deaths were reported. The common grade 3 or greater AEs were leucopenia (18.0%) and neutropenia (16.0%). Five (10%) patients discontinued treatment because of AEs. CONCLUSION Rh-endostatin plus irinotecan/cisplatin showed promising anti-tumor activity in advanced ESCC patients with a good safety profile in the second-line setting, which warrants further study in this population. (ClinicalTrials.gov identifier: NCT03797625).
Collapse
Affiliation(s)
- Zhihuang Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Institute of Thoracic Oncology, Fudan University, Shanghai, People’s Republic of China
| | - Si Sun
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Institute of Thoracic Oncology, Fudan University, Shanghai, People’s Republic of China
| | - Xinmin Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Institute of Thoracic Oncology, Fudan University, Shanghai, People’s Republic of China
| | - Hui Yu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Institute of Thoracic Oncology, Fudan University, Shanghai, People’s Republic of China
| | - Xianghua Wu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Institute of Thoracic Oncology, Fudan University, Shanghai, People’s Republic of China
| | - Jialei Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Institute of Thoracic Oncology, Fudan University, Shanghai, People’s Republic of China
| | - Jianhua Chang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, People’s Republic of China
| | - Huijie Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Institute of Thoracic Oncology, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
165
|
Shoari A, Tahmasebi M, Khodabakhsh F, Cohan RA, Oghalaie A, Behdani M. Angiogenic biomolecules specific nanobodies application in cancer imaging and therapy; review and updates. Int Immunopharmacol 2022; 105:108585. [DOI: 10.1016/j.intimp.2022.108585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/05/2022]
|
166
|
Rodoplu D, Matahum JS, Hsu CH. A microfluidic hanging drop-based spheroid co-culture platform for probing tumor angiogenesis. LAB ON A CHIP 2022; 22:1275-1285. [PMID: 35191460 DOI: 10.1039/d1lc01177d] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Co-culturing of embryoid bodies (EBs) and tumor spheroids (TSs) allows mimicking tumor angiogenesis in vitro. Here, we report a microfluidic hanging drop-based spheroid co-culture device (μ-CCD) that permits the generation and co-culturing of EBs and TSs using a simple manual operation procedure and setup. In brief, uniform-sized EBs and TSs can be generated on the device in eight pairs of hanging droplets from adjacent microfluidic channels, followed by the confrontation of EB and TS pairs by merging the droplet pairs to culture the EB-TS spheroids to investigate tumor-induced angiogenic sprouting. The physical parameters of the device were optimized to maintain the long-term stability of hanging droplets for up to ten days. The mouse embryonic stem cell line ES-D3 and breast cancer cell lines MDA-MB-231 and MCF-7 were used to generate EBs, invasive TSs, and non-invasive TSs respectively. Confocal imaging results showed that the vessel percentage area and total vessel length which are linked to tumor angiogenesis increased after 6 days of co-culturing. An anti-angiogenesis drug testing on the co-cultured EB-TS spheroids was also demonstrated in the device. The μ-CCD provides a simple yet high-efficiency method to generate and co-culture cell spheroids and may also be useful for other applications involving spheroid co-culturing.
Collapse
Affiliation(s)
- Didem Rodoplu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan.
| | - Jefunnie Sierra Matahum
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan.
| | - Chia-Hsien Hsu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan.
- Institute of Nanoengineering and Microsystems, National Tsing Hua University, Hsinchu 30013, Taiwan
- Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
167
|
Wu S, Lu L, Zhou J, Ran D, Wang S, Xu Q, Xu W, Wang J, Liu Y, Xie C, Luo Z, Lu W. All-stage targeted therapy for glioblastoma based on lipid membrane coated cabazitaxel nanocrystals. J Control Release 2022; 345:685-695. [PMID: 35346767 DOI: 10.1016/j.jconrel.2022.03.047] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 01/14/2023]
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor with poor prognosis and frequent recurrence. The blood-brain barrier (BBB), blood-brain tumor barrier (BBTB) hinder the entry of therapeutics into the glioma region. Vasculogenic mimicry (VM) formed by invasive glioma cells is also related to recurrence of GBM. VAP is a D-peptide ligand of GRP78 protein overexpressed on BBTB, VM, and glioma cells but not on normal tissues. Besides, p-hydroxybenzoic acid (pHA) can effectively traverse the BBB. Herein we developed an all-stage glioma-targeted cabazitaxel (CBZ) nanocrystal loaded liposome modified with a "Y" shaped targeting ligand composed of pHA and VAP (pV-Lip/cNC). The pure drug nanocrystal core provided high drug loading, while lipid membrane promoted the stability and circulation time. pV-Lip/cNC exhibited excellent glioma homing, barriers crossing, and tumor spheroid penetrating capability in vitro. Treatment of pV-Lip/cNC displayed enhanced CBZ accumulation in glioma and anti-glioma effect with a median survival time (53 days) significantly longer than that of cNC loaded liposomes modified with either single ligand (42 days for VAP and 45 days for pHA) in the murine orthotopic GBM model. These results indicated pV-Lip/cNC could traverse the BBB and BBTB, destruct VM, and finally kill glioma cells to realize all-stage glioma therapy.
Collapse
Affiliation(s)
- Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Linwei Lu
- The Department of Integrative Medicine, Huashan Hospital, Fudan University, and The Institutes of Integrative Medicine of Fudan University, Shanghai 200040, China
| | - Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Danni Ran
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Songli Wang
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Weixia Xu
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; The Department of Integrative Medicine, Huashan Hospital, Fudan University, and The Institutes of Integrative Medicine of Fudan University, Shanghai 200040, China; Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China; Minhang Branch, Zhongshan Hospital and Institute of Fudan-Minghang Academic Health System, Minghang Hospital, Fudan University, Shanghai 201199, China.
| |
Collapse
|
168
|
Germain N, Dhayer M, Dekiouk S, Marchetti P. Current Advances in 3D Bioprinting for Cancer Modeling and Personalized Medicine. Int J Mol Sci 2022; 23:3432. [PMID: 35408789 PMCID: PMC8998835 DOI: 10.3390/ijms23073432] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Tumor cells evolve in a complex and heterogeneous environment composed of different cell types and an extracellular matrix. Current 2D culture methods are very limited in their ability to mimic the cancer cell environment. In recent years, various 3D models of cancer cells have been developed, notably in the form of spheroids/organoids, using scaffold or cancer-on-chip devices. However, these models have the disadvantage of not being able to precisely control the organization of multiple cell types in complex architecture and are sometimes not very reproducible in their production, and this is especially true for spheroids. Three-dimensional bioprinting can produce complex, multi-cellular, and reproducible constructs in which the matrix composition and rigidity can be adapted locally or globally to the tumor model studied. For these reasons, 3D bioprinting seems to be the technique of choice to mimic the tumor microenvironment in vivo as closely as possible. In this review, we discuss different 3D-bioprinting technologies, including bioinks and crosslinkers that can be used for in vitro cancer models and the techniques used to study cells grown in hydrogels; finally, we provide some applications of bioprinted cancer models.
Collapse
Affiliation(s)
- Nicolas Germain
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| | - Melanie Dhayer
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Salim Dekiouk
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Philippe Marchetti
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| |
Collapse
|
169
|
Relationship between VEGF Family Members, Their Receptors and Cell Death in the Neoplastic Transformation of Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23063375. [PMID: 35328794 PMCID: PMC8952321 DOI: 10.3390/ijms23063375] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is the second most common cause of cancer death in the world. Both modifiable and nonmodifiable risk factors play a significant role in the pathogenesis of this tumor. The diagnosis is usually made late due to limitations of screening tests; therefore, the scientists are looking for new diagnostic tools such as gene or miRNA expression or different proteins’ concentrations, e.g., vascular endothelial growth factor (VEGF) family members. The VEGF family (VEGF-A, VEGF-B, VEGF-C, VEGF-D and PlGF) plays a key role in the processes of blood vessel formation in embryonic development as well as in pathological angiogenesis and lymphangiogenesis, which allow the tumor to grow exponentially. Blockage of VEGF-related pathways seems to be a valid therapeutic target. It was suggested in recent studies, that besides already used drugs, e.g., bevacizumab, there are other agents with potential usefulness in anticancer activity such as miRNAs, TMEA, granzyme K, baicalein and arginine. Moreover, VEGF proteins were assessed to induce the expression of anti-apoptotic proteins such as BCL-2 and BAX. Therefore, investigations concerning the usefulness of VEGF family members, not only in the development but also in the therapy of CRC, in order to fully elucidate their role in carcinogenesis, are extremely important.
Collapse
|
170
|
Chibani H, El Bairi K, Al Jarroudi O, Afqir S. Bevacizumab in metastatic colorectal cancer in a real-life setting - toxicity profile, survival outcomes, and impact of tumor sidedness. Contemp Oncol (Pozn) 2022; 26:32-39. [PMID: 35506031 PMCID: PMC9052347 DOI: 10.5114/wo.2022.114678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Targeting angiogenesis in metastatic colorectal cancer (mCRC) using bevacizumab is a standard of care. The addition of this targeted biological agent to first-line infusional fluoropyrimidine-based chemotherapy was associated with superior overall survival (OS) in several randomized and controlled studies for CRC patients in the metastatic setting. However, access to this therapy in countries with limited resources is challenging. In Morocco, bevacizumab was introduced for this indication after considerable efforts of the Ministry of Health and Lalla Salma Foundation to support cancer patients with a limited income. In this report, the real-world efficacy and safety of the combination of bevacizumab with chemotherapy in mCRC are reported based on a retrospective cohort in Eastern Morocco. MATERIAL AND METHODS The archives of the medical records of 98 mCRC patients treated with first-line bevacizumab at the Hassan II Regional Cancer Center (Oujda, Morocco) were sampled from 1st January 2014 to 31st December 2019 and analyzed using descriptive statistics, Kaplan-Meier estimation, and a multivariable Cox regression model for a time-to-event study. RESULTS The toxicity profile was dominated by grade I-II proteinuria (10%), bleeding events (10%), thromboembolic events (9%), grade I-III hypertension (3%), and other rare events such as delayed healing of the stoma, scar dehiscence, intestinal perforation, and heart failure deterioration. In terms of survival, median OS and progression-free survival in the whole cohort were 22 and 13 months respectively. Patients who benefited from a metastasectomy after bevacizumab treatment had 31 months of median OS as compared to 14 months in the matched cohort with non-resectable liver metastasis. Notably, we demonstrated that tumor sidedness is a predictive factor of OS [hazard ratio (HR) = 2.452; 95% CI: 1.434-4.191, p = 0.001]. Moreover, the median OS for patients who received between 10 and 20 or more than 20 bevacizumab administrations was 24 and 33 months respectively as compared to those who received less than 10 cures (17 months) (log rank p < 0.0001). These markedly improved outcomes were also confirmed in multivariate Cox regression. A highly significant association of bevacizumab use and OS was found after adjusting for covariates (HR = 0.518, 95% CI: 0.374-0.717; p < 0.0001). CONCLUSIONS The current study confirmed the important place of this therapeutic strategy in mCRC. Additional studies with prospective enrollment are awaited to validate these findings.
Collapse
Affiliation(s)
- Hind Chibani
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
| | - Khalid El Bairi
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
| | - Ouissam Al Jarroudi
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
| | - Said Afqir
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
| |
Collapse
|
171
|
Youssef ASED, Abdel-Fattah MA, Lotfy MM, Nassar A, Abouelhoda M, Touny AO, Hassan ZK, Mohey Eldin M, Bahnassy AA, Khaled H, Zekri ARN. Multigene Panel Sequencing Reveals Cancer-Specific and Common Somatic Mutations in Colorectal Cancer Patients: An Egyptian Experience. Curr Issues Mol Biol 2022; 44:1332-1352. [PMID: 35723313 PMCID: PMC8947625 DOI: 10.3390/cimb44030090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/11/2022] Open
Abstract
This study aims at identifying common pathogenic somatic mutations at different stages of colorectal carcinogenesis in Egyptian patients. Our cohort included colonoscopic biopsies collected from 120 patients: 20 biopsies from patients with inflammatory bowel disease, 38 from colonic polyp patients, and 62 from patients with colorectal cancer. On top of this, the cohort included 20 biopsies from patients with non-specific mild to moderated colitis. Targeted DNA sequencing using a customized gene panel of 96 colorectal related genes running on the Ion Torrent NGS technology was used to process the samples. Our results revealed that 69% of all cases harbored at least one somatic mutation. Fifty-seven genes were found to carry 232 somatic non-synonymous variants. The most frequently pathogenic somatic mutations were localized in TP53, APC, KRAS, and PIK3CA. In total, 16 somatic mutations were detected in the CRC group and in either the IBD or CP group. In addition, our data showed that 51% of total somatic variants were CRC-specific variants. The average number of CRC-specific variants per sample is 2.4. The top genes carrying CRC-specific mutations are APC, TP53, PIK3CA, FBXW7, ATM, and SMAD4. It seems obvious that TP53 and APC genes were the most affected genes with somatic mutations in all groups. Of interest, 85% and 28% of the APC and TP53 deleterious somatic mutations were located in Exon 14 and Exon 3, respectively. Besides, 37% and 28% of the total somatic mutations identified in APC and TP53 were CRC-specific variants, respectively. Moreover, we identified that, in 29 somatic mutations in 21 genes, their association with CRC patients was unprecedented. Ten detected variants were likely to be novel: six in PIK3CA and four variants in FBXW7. The detected P53, Wnt/βcatenin, Angiogenesis, EGFR, TGF-β and Interleukin signaling pathways were the most altered pathways in 22%, 16%, 12%, 10%, 9% and 9% of the CRC patients, respectively. These results would contribute to a better understanding of the colorectal cancer and in introducing personalized therapies for Egyptian CRC patients.
Collapse
Affiliation(s)
- Amira Salah El-Din Youssef
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| | | | - Mai M. Lotfy
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| | - Auhood Nassar
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| | | | - Ahmed O. Touny
- Surgical Oncology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Zeinab K. Hassan
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| | - Mohammed Mohey Eldin
- Tropical Medicine Department, El Kasr Al-Aini, Cairo University, Cairo 11562, Egypt;
| | - Abeer A. Bahnassy
- Molecular Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Hussein Khaled
- Medical Oncology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Abdel Rahman N. Zekri
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| |
Collapse
|
172
|
Su S, Chen W, Zheng M, Lu G, Tang W, Huang H, Qu D. Facile Fabrication of 3D-Printed Porous Ti6Al4V Scaffolds with a Sr-CaP Coating for Bone Regeneration. ACS OMEGA 2022; 7:8391-8402. [PMID: 35309469 PMCID: PMC8928158 DOI: 10.1021/acsomega.1c05908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/10/2022] [Indexed: 05/12/2023]
Abstract
To improve osseointegration caused by the stress-shielding effect and the inert nature of titanium-based alloys, in this work, we successfully constructed a strontium calcium phosphate (Sr-CaP) coating on three-dimensional (3D)-printed Ti6Al4V scaffolds to address this issue. The energy-dispersive X-ray spectroscopy (EDS) and X-ray diffraction (XRD) results indicated that the coatings with and without Sr doping mainly consisted of CaHPO4. The bonding strength of Sr doping coating met the required ISO 13 779-4-2018 standard (≥15 MPa). The in vitro results suggested that the Sr-CaP-modified Ti6Al4V scaffolds were found to effectively promote mice bone-marrow stem cell (mBMSC) adhesion, spreading, and osteogenesis. The in vivo experiments also showed that the Sr-CaP-modified Ti6Al4V scaffolds could significantly improve bone regeneration and osseointegration. More importantly, Sr-doped CaP-coated Ti6Al4V scaffolds were found to accelerate bone healing in comparison to CaP-coated Ti6Al4V scaffolds. The Sr-CaP-modified Ti6Al4V scaffolds are considered a promising strategy to develop bioactive surfaces for enhancing the osseointegration between the implant and bone tissue.
Collapse
Affiliation(s)
- Shenghui Su
- Division
of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Weidong Chen
- Division
of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Minghui Zheng
- Division
of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
- Department
of Orthopaedic Surgery, Zengcheng Branch of Nanfang Hospital, Southern Medical University, 511338 Guangzhou, China
| | - Guozan Lu
- Guangzhou
Huatai 3D Material Manufacture Ltd., Co., 511300 Guangzhou, China
| | - Wei Tang
- Department
of Anatomy, College of Basic Medicine, Dalian
Medical University, Dalian 116044, China
| | - Haihong Huang
- Division
of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Dongbin Qu
- Division
of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
- Department
of Orthopaedic Surgery, Zengcheng Branch of Nanfang Hospital, Southern Medical University, 511338 Guangzhou, China
| |
Collapse
|
173
|
Inverting angiogenesis with interstitial flow and chemokine matrix-binding affinity. Sci Rep 2022; 12:4237. [PMID: 35273299 PMCID: PMC8913640 DOI: 10.1038/s41598-022-08186-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/15/2022] [Indexed: 11/30/2022] Open
Abstract
The molecular signaling pathways that orchestrate angiogenesis have been widely studied, but the role of biophysical cues has received less attention. Interstitial flow is unavoidable in vivo, and has been shown to dramatically change the neovascular patterns, but the mechanisms by which flow regulates angiogenesis remain poorly understood. Here, we study the complex interactions between interstitial flow and the affinity for matrix binding of different chemokine isoforms. Using a computational model, we find that changing the matrix affinity of the chemokine isoform can invert the effect of interstitial flow on angiogenesis—from preferential growth in the direction of the flow when the chemokine is initially matrix-bound to preferential flow against the flow when it is unbound. Although fluid forces signal endothelial cells directly, our data suggests a mechanism for the inversion based on biotransport arguments only, and offers a potential explanation for experimental results in which interstitial flow produced preferential vessel growth with and against the flow. Our results point to a particularly intricate effect of interstitial flow on angiogenesis in the tumor microenvironment, where the vessel network geometry and the interstitial flow patterns are complex.
Collapse
|
174
|
Zhao L, Wang YF, Liu J, Jiang BH, Liu LZ. Human endothelial cells promote arsenic-transformed lung epithelial cells to induce tumor growth and angiogenesis through interleukin-8 induction. Aging (Albany NY) 2022; 14:2113-2130. [PMID: 35241635 PMCID: PMC8954972 DOI: 10.18632/aging.203930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/29/2022] [Indexed: 11/25/2022]
Abstract
Arsenic exposure is associated with lung cancer. Angiogenesis is essential for tumor development. However, the role and mechanism of human vascular endothelial cells in tumor growth and angiogenesis induced by arsenic-transformed bronchial epithelial (As-T) cells remain to be elucidated. In this study, we found that endothelial cells significantly increased As-T cell-induced tumor growth compared to those induced by As-T cells alone. To understand the molecular mechanism, we found that endothelial cells co-cultured with As-T cells or cultured in conditioned medium (CM) prepared from As-T cells showed much higher cell migration, proliferation, and tube formation compared to those co-cultured with BEAS-2B (B2B) cells or cultured in CM from B2B. We identified that higher levels of intracellular interleukin 8 (IL-8) were secreted by As-T cells, which activated IL-8/IL-8R signaling to promote endothelial cells migration and tube formation. IL-8 silencing and knockout (KO) in As-T cells, or IL-8 neutralizing antibody dramatically suppressed endothelial cell proliferation, migration, tube formation in vitro, and tumor growth and angiogenesis in vivo, suggesting a key role of IL-8 in As-T cells to induce angiogenesis via a paracrine effect. Finally, blocking of IL-8 receptors C-X-C chemokine receptor type 1 (CXCR1) and CXCR2 with neutralizing antibodies and chemical inhibitors inhibited tube formation, indicating that IL-8Rs on endothelial cells are necessary for As-T cell-induced angiogenesis. Overall, this study reveals an important molecular mechanism of arsenic-induced carcinogenesis, and suggests a new option to prevent and treat arsenic-induced angiogenesis.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yi-Fang Wang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jie Liu
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bing-Hua Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ling-Zhi Liu
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
175
|
Jyothi M, Banumathi, Zabiulla, Sherapura A, Khamees HA, Prabhakar B, Khanum SA. Synthesis, structure analysis, DFT calculations and energy frameworks of new coumarin appended oxadiazoles, to regress ascites malignancy by targeting VEGF mediated angiogenesis. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
176
|
Abdalla A, Murali C, Amin A. Safranal Inhibits Angiogenesis via Targeting HIF-1α/VEGF Machinery: In Vitro and Ex Vivo Insights. Front Oncol 2022; 11:789172. [PMID: 35211395 PMCID: PMC8862147 DOI: 10.3389/fonc.2021.789172] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/30/2021] [Indexed: 12/27/2022] Open
Abstract
Nature has a nearly infinite inventory of unexplored phytochemicals and biomolecules that have the potential to treat a variety of diseases. Safranal exhibits anti-cancer property and the present study explores its antiangiogenic property. Hepatocellular carcinoma (HCC) ranks as the sixth deadliest among all cancer types. Targeting the non-tumor vasculature supporting system is very promising as it has less plasticity, unlike malignant cells that are often associated with issues like drug resistance, poor prognosis, and relapse. In this study, we successfully inhibited the proliferation of primary human umbilical vein endothelial cells (HUVEC) with an IC50 of 300μM and blocked VEGF secretion in HepG2 cells. Furthermore, safranal inhibited VEGF-induced angiogenesis in vitro and ex vivo via scratch wound assay, tube formation assay, transmembrane assay, and aortic ring assay. In addition, safranal downregulated the in vitro expression of HIF-1α, VEGF, VEGFR2, p-AKT, p-ERK1/2, MMP9, p-FAK, and p-STAT3. The present study is the first to reveal the antiangiogenic potential of safranal and propose its possible underlying mechanism in HCC.
Collapse
Affiliation(s)
- Ali Abdalla
- Weinberg Institute for Cognitive Science, University of Michigan, Ann Arbor, MI, United States
| | - Chandraprabha Murali
- Biology Department, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Amr Amin
- Biology Department, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates.,The College, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
177
|
Nafar S, Nouri N, Alipour M, Fallahi J, Zare F, Tabei SMB. Exosome as a target for cancer treatment. J Investig Med 2022; 70:1212-1218. [PMID: 35210328 DOI: 10.1136/jim-2021-002194] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 11/03/2022]
Abstract
Exosomes are small vesicles covered by a lipid bilayer, ranging in size from 50 nm to 90 nm, secreted by different cell types in the body under normal and pathological conditions. They are surrounded by cell-segregated membrane complexes and play a role in the pathological and physiological environments of target cells by transfer of different molecules such as microRNA (miRNA). Exosomes have been detected in many body fluids, such as in the amniotic fluid, urine, breast milk, blood, saliva, ascites, semen, and bile. They include proteins, lipids, and nucleic acids such as DNA, RNA, and miRNA, which have many functions in target cells under pathological and physiological conditions. They participate in pathological processes such as tumor growth and survival, autoimmunity, neurodegenerative disorders, infectious diseases, inflammation conditions, and others. Biomarkers in exosomes isolated from body fluids have allowed for a more precise and consistent diagnostic method than previous approaches. Exosomes can be used in a variety of intracellular functions, and with advances in molecular techniques they can be used in the treatment and diagnosis of many diseases, including cancer. These vesicles play a significant role in various stages of cancer. Tumor-derived exosomes have an important role in tumor growth, survival, and metastasis. In contrast, the use of stem cells in cancer treatment is a relatively new scientific area. We hope to address targeted use of miRNA-carrying exosomes in cancer therapy in this review paper.
Collapse
Affiliation(s)
- Samira Nafar
- Department of Genetics, Shiraz University of Medical Science, Shiraz, Iran
| | - Negar Nouri
- Student Research Committee, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Maedeh Alipour
- MSc of Hematology and Blood Bank, Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fateme Zare
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | |
Collapse
|
178
|
Bibak B, Shakeri F, Keshavarzi Z, Mollazadeh H, Javid H, Jalili-Nik M, Sathyapalan T, Afshari AR, Sahebkar A. Anticancer mechanisms of Berberine: a good choice for glioblastoma multiforme therapy. Curr Med Chem 2022; 29:4507-4528. [PMID: 35209812 DOI: 10.2174/0929867329666220224112811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 12/30/2021] [Accepted: 01/05/2022] [Indexed: 11/22/2022]
Abstract
The most typical malignant brain tumor, glioblastoma multiforme (GBM), seems to have a grim outcome, despite the intensive multi-modality interventions. Literature suggests that biologically active phytomolecules may exert anticancer properties by regulating several signaling pathways. Berberine, an isoquinoline alkaloid, has various pharmacological applications to combat severe diseases like cancer. Mechanistically, Berberine inhibits cell proliferation and invasion, suppresses tumor angiogenesis, and induces cell apoptosis. The effect of the antitumoral effect of Berberine in GBM is increasingly recognized. This review sheds new light on the regulatory signaling mechanisms of Berberine in various cancer, proposing its potential role as a therapeutic agent for GBM. .
Collapse
Affiliation(s)
- Bahram Bibak
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Farzaneh Shakeri
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zakieh Keshavarzi
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hamid Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hossein Javid
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Jalili-Nik
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
179
|
Gharib AF, Elsawy WH, Alrehaili AA, Amin HS, Alhuthali HM, Bakhuraysah MM, El Askary A. The Application of Molecular Techniques for Assessment of SOX2 and miR126 Expression as Prognostic Markers in Esophageal Carcinoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:1514412. [PMID: 39290848 PMCID: PMC11407893 DOI: 10.1155/2022/1514412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/27/2022] [Accepted: 02/10/2022] [Indexed: 09/19/2024]
Abstract
Objective To study the problem in esophageal cancer, the function of SOX2 and miR-126 has not been completely explored. The objective of this study was to find out how SOX2 and miR-126 act in esophageal cancer and their relation to the clinical and prognostic features. Methods The expression of SOX2 and miR-126 was properly assessed in the carcinoma of the esophagus, and the nearby healthy tissues surgically excised from 35 included patients. Results SOX2 was elevated in esophageal cancer relative to normal tissues contrary to the miR-126 levels. This inverse relationship was linked to adverse clinical features. Background SOX2 has been involved as an oncogene in various types of malignant tumors; microRNA-126 (miR-126) is extensively expressed in vascular endothelial cells, which control angiogenesis. Furthermore, many published reports reasonably concluded that based on the prime characteristic of malignant cells, miR-126 may act appropriately as a promotor or a suppressor for the malignant growth. Conclusion In esophageal cancer, SOX2 works as an oncogene, whereas miR-126 acts as a tumor suppressor gene. SOX2 overexpression and miR-126 downregulation were shown to be linked to a poor prognosis.
Collapse
Affiliation(s)
- Amal F Gharib
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif 21944, Saudi Arabia
| | - Wael H Elsawy
- Department of Clinical Oncology, Faculty of Medicine, Zagazig University, Egypt
| | - Amani A Alrehaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif 21944, Saudi Arabia
| | - Hanan S Amin
- Department of Clinical Chemistry, Theodor Bilharz Research Institute, Cairo, Egypt
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif 21944, Saudi Arabia
| | - Maha M Bakhuraysah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif 21944, Saudi Arabia
| | - Ahmad El Askary
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
180
|
Poto R, Cristinziano L, Modestino L, de Paulis A, Marone G, Loffredo S, Galdiero MR, Varricchi G. Neutrophil Extracellular Traps, Angiogenesis and Cancer. Biomedicines 2022; 10:biomedicines10020431. [PMID: 35203640 PMCID: PMC8962440 DOI: 10.3390/biomedicines10020431] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/07/2022] Open
Abstract
Human neutrophils, the most abundant circulating leukocytes, are fundamental components of the host response against different pathogens. Until a few years ago, neutrophils received limited attention in cancer immunology. Recently, it was discovered that both circulating, and tumor-associated, neutrophils possess functional plasticity when exposed to various inflammatory stimuli and in the tumor microenvironment. Neutrophils and their mediators can exert several pro-tumor activities in cancer and promote metastasis through different mechanisms. Angiogenesis plays a pivotal role in inflammation and tumor growth. Activated human neutrophils release several angiogenic factors [vascular endothelial growth factor-A (VEGF-A), angiopoietin-1 (ANGPT1), CXCL8, hepatocyte growth factor (HGF), and metalloproteinase 9 (MMP-9)] and form neutrophil extracellular traps (NETs). NETs promote tumor growth and metastasis formation through several mechanisms: they can awake dormant cancer cells, capture circulating tumor cells, coat and shield cancer cells, thus preventing CD8+- and natural killer (NK) cell-mediated cytotoxicity. ANGPTs released by endothelial and periendothelial mural cells induce platelet-activating factor (PAF) synthesis and neutrophil adhesion to endothelial cells. NETs can directly exert several proangiogenic activities in human endothelial cells and NETs induced by ANGPTs and PAF increase several aspects of angiogenesis in vitro and in vivo. A better understanding of the pathophysiological functions of NETs in cancer and angiogenesis could be of importance in the early diagnosis, prevention and treatment of tumors.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (L.C.); (L.M.); (A.d.P.); (G.M.); (S.L.); (M.R.G.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Leonardo Cristinziano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (L.C.); (L.M.); (A.d.P.); (G.M.); (S.L.); (M.R.G.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Luca Modestino
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (L.C.); (L.M.); (A.d.P.); (G.M.); (S.L.); (M.R.G.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (L.C.); (L.M.); (A.d.P.); (G.M.); (S.L.); (M.R.G.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (L.C.); (L.M.); (A.d.P.); (G.M.); (S.L.); (M.R.G.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (L.C.); (L.M.); (A.d.P.); (G.M.); (S.L.); (M.R.G.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (L.C.); (L.M.); (A.d.P.); (G.M.); (S.L.); (M.R.G.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (L.C.); (L.M.); (A.d.P.); (G.M.); (S.L.); (M.R.G.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
181
|
Van Petten de Vasconcelos Azevedo F, Lopes DS, Zóia MAP, Correia LIV, Saito N, Fonseca BB, Polloni L, Teixeira SC, Goulart LR, de Melo Rodrigues Ávila V. A New Approach to Inhibiting Triple-Negative Breast Cancer: In Vitro, Ex Vivo and In Vivo Antiangiogenic Effect of BthTx-II, a PLA 2-Asp-49 from Bothrops jararacussu Venom. Biomolecules 2022; 12:258. [PMID: 35204758 PMCID: PMC8961627 DOI: 10.3390/biom12020258] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 12/10/2022] Open
Abstract
Phospholipases A2 (PLA2) represent a superfamily of enzymes widely distributed in living organisms, with a broad spectrum of pharmacological activities and therapeutic potential. Anti-angiogenic strategies have become one of the main tools in fighting cancer. In this sense, the present work reports the inhibition of tumor angiogenesis induced by Asp-49 BthTX-II using in vitro, ex vivo and in vivo approaches. We demonstrate that BthTx-II inhibited cell adhesion, proliferation, and migration of human umbilical vein endothelial cells (HUVEC), as well as caused a reduction in the levels of endothelial growth factor (VEGF) during in vitro angiogenesis assays. BthTx-II was also able to inhibit the sprouting angiogenic process, by the ex vivo germination assay of the aortic ring; in addition, this toxin inhibited the migration and proliferation of HUVEC in co-culture with triple-negative breast cancer cells (e.g., MDA-MB-231 cells). Finally, in vivo tumor suppression and anti-angiogenic activities were analyzed using MDA-MB-231 cells with Matrigel injected into the chorioallantoic membrane of chicken embryo (CAM) for 7 days treatment with BthTx-II, showing a considerable reduction in vessel caliber, on the size and weight of tumors. Together, these results suggest an important antiangiogenic and antitumor role for BthTx-II, as a potential prototype for the development of new tools and antitumor drugs in cancer therapy.
Collapse
Affiliation(s)
- Fernanda Van Petten de Vasconcelos Azevedo
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (L.I.V.C.); (L.P.)
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | - Daiana Silva Lopes
- Multidisciplinary Institute of Health, Federal University of Bahia, Vitoria da Conquista, Salvador 40170-110, BA, Brazil;
| | - Mariana Alves Pereira Zóia
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | - Lucas Ian Veloso Correia
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (L.I.V.C.); (L.P.)
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | - Natieli Saito
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | | | - Lorena Polloni
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (L.I.V.C.); (L.P.)
| | - Samuel Cota Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil;
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | - Veridiana de Melo Rodrigues Ávila
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (L.I.V.C.); (L.P.)
| |
Collapse
|
182
|
CREB3L4 promotes angiogenesis and tumor progression in gastric cancer through regulating VEGFA expression. Cancer Gene Ther 2022; 29:241-252. [PMID: 33637885 DOI: 10.1038/s41417-021-00305-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/21/2021] [Accepted: 02/02/2021] [Indexed: 01/30/2023]
Abstract
Tumor angiogenesis is a key step in the progression of gastric cancer (GC) that delivers essential nutrients and oxygen to tumor cells and distant sites. The cyclic AMP responsive element-binding protein 3-like 4 (CREB3L4) is a transcription factor highly expressed in multiple human cancers. This study aimed to investigate the regulatory effects of CREB3L4 on GC progression and angiogenesis. CREB3L4 was overexpressed in GC tissues and cell lines, and was positively correlated with advanced tumor stage and poor survival in GC patients. The upregulation of CREB3L4 in GC cells increased cell viability, promoted cell proliferation, reduced apoptosis, enhanced cell migration and invasion, and induced the formation of tubule-like endothelial structures, whereas CREB3L4 knockdown impeded tumor cell growth, attenuated cell motility, and prevented human umbilical vein endothelial cells from forming tubule-like structures. In addition, mice inoculated with CREB3L4-deficient GC cells showed significantly suppressed tumor growth compared to the group harboring wild-type tumors. Further analysis revealed that CREB3L4 expression was positively correlated with the level of vascular endothelial growth factor A (VEGFA) in gastric tumors. CREB3L4 regulated the transcription activity of VEGFA by binding to its promoter. The downregulation of VEGFA eliminated CREB3L4-induced GC cell growth and movement, and the formation of endothelial structures; while VEGFA upregulation greatly induced the growth and movement of GC cells with CREB3L4 deficiency. In conclusion, CREB3L4 promoted gastric tumor progression and endothelial angiogenesis by transcriptionally activating the VEGFA promoter, suggesting that therapeutic potential of the CREB3L4/VEGFA axis in GC treatment.
Collapse
|
183
|
de Abrantes RA, Batista TM, Mangueira VM, de Sousa TKG, Ferreira RC, Moura APG, Abreu LS, Alves AF, Velozo ES, Batista LM, da Silva MS, Tavares JF, Sobral MV. Antitumor and antiangiogenic effects of Tonantzitlolone B, an uncommon diterpene from Stillingia loranthacea. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:267-274. [PMID: 34854946 DOI: 10.1007/s00210-021-02185-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/14/2021] [Indexed: 01/04/2023]
Abstract
Natural products have played a pivotal role for the discovery of anticancer drugs. Tonantzitlolones are flexibilan-type diterpenes rare in nature; therefore, few reports have shown antiviral and cytotoxic activities. This study aimed to investigate the in vivo antitumor action of Tonantzitlolone B (TNZ-B) and its toxicity. Toxicity was evaluated in mice (acute and micronucleus assays). Antitumor activity of TNZ-B (1.5 or 3 mg/kg intraperitoneally - i.p.) was assessed in Ehrlich ascites carcinoma model. Angiogenesis and reactive oxygen species (ROS) and nitric oxide (NO) production were also investigated, in addition to toxicological effects after 7-day treatment. The LD50 (lethal dose 50%) was estimated at around 25 mg/kg (i.p.), and no genotoxicity was recorded. TNZ-B reduced the Ehrlich tumor's volume and total viable cancer cell count (p < 0.001 for both). Additionally, TNZ-B reduced peritumoral microvessel density (p < 0.01), suggesting antiangiogenic action. Moreover, a decrease was observed on ROS (p < 0.05) and nitric oxide (p < 0.001) levels. No significant clinical findings were observed in the analysis of biochemical, hematological, and histological (liver and kidney) parameters. In conclusion, TNZ-B exerts antitumor and antiangiogenic effects by reducing ROS and NO levels and has weak in vivo dose-repeated toxicity. These data contribute to elucidate the antitumor action of TNZ-B and point the way for further studies with this natural compound as an anticancer drug.
Collapse
Affiliation(s)
- Renata A de Abrantes
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Tatianne M Batista
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Vivianne M Mangueira
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Tatyanna K G de Sousa
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Rafael C Ferreira
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Ana Paula G Moura
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Lucas S Abreu
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Adriano F Alves
- Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Eudes S Velozo
- Research Laboratory in Materia Medica, School of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Leônia M Batista
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Brazil
| | - Marcelo S da Silva
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Brazil
| | - Josean F Tavares
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Brazil
| | - Marianna V Sobral
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil.
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Brazil.
| |
Collapse
|
184
|
Systemic Treatment of Ewing Sarcoma: Current Options and Future Perspectives. FORUM OF CLINICAL ONCOLOGY 2022. [DOI: 10.2478/fco-2021-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Ewing sarcoma (ES) is an uncommon malignant neoplasm, mostly affecting young adults and adolescents. Surgical excision, irradiation, and combinations of multiple chemotherapeutic agents are currently used as a multimodal strategy for the treatment of local and oligometastatic disease. Although ES usually responds to the primary treatment, relapsed and primarily refractory disease remains a difficult therapeutic challenge. The growing understanding of cancer biology and the subsequent development of new therapeutic strategies have been put at the service of research in recurrent and refractory ES, generating a great number of ongoing studies with compounds that could find superior clinical outcomes in the years to come. This review gathers the current available information on the treatment and clinical investigation of ES and aims to be a point of support for future research.
Collapse
|
185
|
Design and Synthesis of New 2-Oxoquinoxalinyl-1,2,4-triazoles as Antitumor VEGFR-2 Inhibitors. Bioorg Chem 2022; 121:105696. [DOI: 10.1016/j.bioorg.2022.105696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/27/2022] [Accepted: 02/17/2022] [Indexed: 01/01/2023]
|
186
|
Fu J, Yu M, Xu W, Yu S. Research Progress of Bile Acids in Cancer. Front Oncol 2022; 11:778258. [PMID: 35127481 PMCID: PMC8810494 DOI: 10.3389/fonc.2021.778258] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/27/2021] [Indexed: 01/09/2023] Open
Abstract
Bile acids (BAs) were originally known as detergents to facilitate the digestion and absorption of lipids. And our current knowledge of BAs has been extended to potential carcinogenic or cancer suppressor factors due to constant research. In fact, BAs were regarded as a tumor promoters as early as the 1940s. Differential bile acid signals emitted by various bile acid profiles can produce distinct pathophysiological traits, thereby participating in the occurrence and development of tumors. Nevertheless, in recent years, more and more studies have noticed the value of BAs as therapeutic targets. And several studies have applied BAs as a therapeutic agent for various diseases including cancer. Based on the above evidence, we acknowledge that the role of BAs in cancer has yet to be exploited, although considerable efforts have been made to probe the functions of BAs. In this review, we describe the characteristics of BAs as a double-edged sword in cancer, hoping to provide references for future cancer treatments.
Collapse
Affiliation(s)
- Junhao Fu
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Min Yu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Wenxia Xu
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Shian Yu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- *Correspondence: Shian Yu,
| |
Collapse
|
187
|
Ruzic D, Djoković N, Srdić-Rajić T, Echeverria C, Nikolic K, Santibanez JF. Targeting Histone Deacetylases: Opportunities for Cancer Treatment and Chemoprevention. Pharmaceutics 2022; 14:pharmaceutics14010209. [PMID: 35057104 PMCID: PMC8778744 DOI: 10.3390/pharmaceutics14010209] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
The dysregulation of gene expression is a critical event involved in all steps of tumorigenesis. Aberrant histone and non-histone acetylation modifications of gene expression due to the abnormal activation of histone deacetylases (HDAC) have been reported in hematologic and solid types of cancer. In this sense, the cancer-associated epigenetic alterations are promising targets for anticancer therapy and chemoprevention. HDAC inhibitors (HDACi) induce histone hyperacetylation within target proteins, altering cell cycle and proliferation, cell differentiation, and the regulation of cell death programs. Over the last three decades, an increasing number of synthetic and naturally derived compounds, such as dietary-derived products, have been demonstrated to act as HDACi and have provided biological and molecular insights with regard to the role of HDAC in cancer. The first part of this review is focused on the biological roles of the Zinc-dependent HDAC family in malignant diseases. Accordingly, the small-molecules and natural products such as HDACi are described in terms of cancer therapy and chemoprevention. Furthermore, structural considerations are included to improve the HDACi selectivity and combinatory potential with other specific targeting agents in bifunctional inhibitors and proteolysis targeting chimeras. Additionally, clinical trials that combine HDACi with current therapies are discussed, which may open new avenues in terms of the feasibility of HDACi’s future clinical applications in precision cancer therapies.
Collapse
Affiliation(s)
- Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.R.); (N.D.); (K.N.)
| | - Nemanja Djoković
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.R.); (N.D.); (K.N.)
| | - Tatjana Srdić-Rajić
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia;
| | - Cesar Echeverria
- Facultad de Medicina, Universidad de Atacama, Copayapu 485, Copiapo 1531772, Chile;
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.R.); (N.D.); (K.N.)
| | - Juan F. Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotica 4, POB 102, 11129 Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370854, Chile
- Correspondence: ; Tel.: +381-11-2685-788; Fax: +381-11-2643-691
| |
Collapse
|
188
|
Genova C, Dellepiane C, Carrega P, Sommariva S, Ferlazzo G, Pronzato P, Gangemi R, Filaci G, Coco S, Croce M. Therapeutic Implications of Tumor Microenvironment in Lung Cancer: Focus on Immune Checkpoint Blockade. Front Immunol 2022; 12:799455. [PMID: 35069581 PMCID: PMC8777268 DOI: 10.3389/fimmu.2021.799455] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
In the last decade, the treatment of non-small cell lung cancer (NSCLC) has been revolutionized by the introduction of immune checkpoint inhibitors (ICI) directed against programmed death protein 1 (PD-1) and its ligand (PD-L1), or cytotoxic T lymphocyte antigen 4 (CTLA-4). In spite of these improvements, some patients do not achieve any benefit from ICI, and inevitably develop resistance to therapy over time. Tumor microenvironment (TME) might influence response to immunotherapy due to its prominent role in the multiple interactions between neoplastic cells and the immune system. Studies investigating lung cancer from the perspective of TME pointed out a complex scenario where tumor angiogenesis, soluble factors, immune suppressive/regulatory elements and cells composing TME itself participate to tumor growth. In this review, we point out the current state of knowledge involving the relationship between tumor cells and the components of TME in NSCLC as well as their interactions with immunotherapy providing an update on novel predictors of benefit from currently employed ICI or new therapeutic targets of investigational agents. In first place, increasing evidence suggests that TME might represent a promising biomarker of sensitivity to ICI, based on the presence of immune-modulating cells, such as Treg, myeloid derived suppressor cells, and tumor associated macrophages, which are known to induce an immunosuppressive environment, poorly responsive to ICI. Consequently, multiple clinical studies have been designed to influence TME towards a pro-immunogenic state and subsequently improve the activity of ICI. Currently, the mostly employed approach relies on the association of "classic" ICI targeting PD-1/PD-L1 and novel agents directed on molecules, such as LAG-3 and TIM-3. To date, some trials have already shown promising results, while a multitude of prospective studies are ongoing, and their results might significantly influence the future approach to cancer immunotherapy.
Collapse
Affiliation(s)
- Carlo Genova
- UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università degli Studi di Genova, Genova, Italy
| | - Chiara Dellepiane
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Paolo Carrega
- Dipartimento di Patologia Umana, University of Messina, Messina, Italy
| | - Sara Sommariva
- SuPerconducting and Other INnovative Materials and Devices Institute, Consiglio Nazionale delle Ricerche (CNR-SPIN), Genova, Italy
- Life Science Computational Laboratory (LISCOMP), IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Guido Ferlazzo
- Dipartimento di Patologia Umana, University of Messina, Messina, Italy
| | - Paolo Pronzato
- UO Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Rosaria Gangemi
- UO Bioterapie, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gilberto Filaci
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università degli Studi di Genova, Genova, Italy
- UO Bioterapie, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Michela Croce
- UO Bioterapie, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
189
|
Abdel Ghany LMA, El-Dydamony NM, Helwa AA, Abdelraouf SM, Abdelnaby RM. Coumarin-acetohydrazide derivatives as novel antiproliferative agents via VEGFR-2/AKT axis inhibition and apoptosis triggering. NEW J CHEM 2022. [DOI: 10.1039/d2nj02436e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The VEGFR-2/AKT pathway is a crucial axis in tumor survival where it is highly dysregulated in many cancer types.
Collapse
Affiliation(s)
- Lina M. A. Abdel Ghany
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Nehad M. El-Dydamony
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Amira A. Helwa
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Sahar M. Abdelraouf
- Biochemistry Department, Faculty of pharmacy, Misr International University, Cairo, Egypt
| | - Rana M. Abdelnaby
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
190
|
Guo SB, Huang WJ, Tian XP. Brusatol modulates diverse cancer hallmarks and signaling pathways as a potential cancer therapeutic. ACTA MATERIA MEDICA 2022; 1. [DOI: 10.15212/amm-2022-0014] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Cancer is a consequence of uncontrolled cell proliferation that is associated with cell-cycle disruption. It is a multifactorial disease that depends on the modulation of numerous oncogenic signaling pathways and targets. Although a battle against cancer has been waged for centuries, this disease remains a major cause of death worldwide. Because of the development of resistance to current anticancer drugs, substantial effort has been focused on discovering more effective agents for tumor therapy. Natural products have powerful prospects as anticancer drugs. Brusatol, a component isolated from the plant Brucea javanica, has been demonstrated to efficiently combat a wide variety of tumors. Extensive studies have indicated that brusatol exhibits anticancer effects by arresting the cell cycle; promoting apoptosis; inducing autophagy; attenuating epithelial-mesenchymal transition; inhibiting migration, invasion and angiogenesis; and increasing chemosensitivity and radiosensitivity. These effects involve various oncogenic signaling pathways, including the MAPK, NF-κB, PI3K/AKT/mTOR, JAK/STAT and Keap1/Nrf2/ARE signaling pathways. This review describes the evidence suggesting that brusatol is a promising drug candidate for cancer therapeutics.
Collapse
Affiliation(s)
- Song-Bin Guo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Wei-Juan Huang
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, P.R. China
| | - Xiao-Peng Tian
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| |
Collapse
|
191
|
Kumar S, Verma R, Tyagi N, Gangenahalli G, Verma YK. Therapeutics effect of mesenchymal stromal cells in reactive oxygen species-induced damages. Hum Cell 2022; 35:37-50. [PMID: 34800267 PMCID: PMC8605474 DOI: 10.1007/s13577-021-00646-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/10/2021] [Indexed: 12/16/2022]
Abstract
Reactive Oxygen Species are chemically unstable molecules generated during aerobic respiration, especially in the electron transport chain. ROS are involved in various biological functions; any imbalance in their standard level results in severe damage, for instance, oxidative damage, inflammation in a cellular system, and cancer. Oxidative damage activates signaling pathways, which result in cell proliferation, oncogenesis, and metastasis. Since the last few decades, mesenchymal stromal cells have been explored as therapeutic agents against various pathologies, such as cardiovascular diseases, acute and chronic kidney disease, neurodegenerative diseases, macular degeneration, and biliary diseases. Recently, the research community has begun developing several anti-tumor drugs, but these therapeutic drugs are ineffective. In this present review, we would like to emphasize MSCs-based targeted therapy against pathologies induced by ROS as cells possess regenerative potential, immunomodulation, and migratory capacity. We have also focused on how MSCs can be used as next-generation drugs with no side effects.
Collapse
Affiliation(s)
- Subodh Kumar
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences (INMAS), Defence Research and Development Organisation (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Ranjan Verma
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences (INMAS), Defence Research and Development Organisation (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Nishant Tyagi
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences (INMAS), Defence Research and Development Organisation (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Gurudutta Gangenahalli
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences (INMAS), Defence Research and Development Organisation (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Yogesh Kumar Verma
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences (INMAS), Defence Research and Development Organisation (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
192
|
Liu YX, Xu BW, Niu XD, Chen YJ, Fu XQ, Wang XQ, Yin CL, Chou JY, Li JK, Wu JY, Bai JX, Wu Y, Li SM, Yu ZL. Inhibition of Src/STAT3 signaling-mediated angiogenesis is involved in the anti-melanoma effects of dioscin. Pharmacol Res 2022; 175:105983. [PMID: 34822972 DOI: 10.1016/j.phrs.2021.105983] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/26/2022]
Abstract
Angiogenesis plays an important role in the growth and metastasis of solid tumors including melanoma. Inhibiting tumor-associated angiogenesis is a tactic in treating melanoma. Dioscin restrains angiogenesis in colon tumor and has anti-melanoma effects in cell and animal models. In a previous study, we found that dioscin inhibits Src/STAT3 signaling in melanoma cells. Activation of the Src/STAT3 pathway has been shown to promote tumor angiogenesis. This study aimed to determine whether dioscin's anti-melanoma effects is related to inhibiting Src/STAT3 signaling-mediated angiogenesis. In a B16F10 allograft mouse model, we found that dioscin inhibited melanoma growth and angiogenesis. To exclude the impact of tumor growth on angiogenesis, a chicken chorioallantoic membrane (CAM) model was used to verify the anti-angiogenic effect of dioscin. Results showed that dioscin suppressed vessel formation in CAM. To determine if tumor secreted pro-angiogenic cytokines are involved in the anti-angiogenic effect of dioscin, conditioned media from dioscin-treated A375 melanoma cells were used to culture human umbilical vein endothelial cells (HUVECs), and tube formation was monitored. It was observed that the tube formation of HUVECs was inhibited. Mechanistic studies revealed that dioscin inhibited the activation of Src and STAT3, and lowered mRNA and protein levels of STAT3 transcriptionally-regulated genes, in B16F10 melanomas. ELISA assays showed that dioscin decreased the secretion of MMP-2, MMP-9 and VEGF from A375 cells. Over-activation of STAT3 lessened the effects of dioscin in decreasing the secretion of pro-angiogenic cytokines from melanoma cells, and in inhibiting tube formation of HUVECs cultured with conditioned media from melanoma cell cultures. In summary, we for the first time demonstrated that inhibiting Src/STAT3 signaling-mediated angiogenesis is involved in the anti-melanoma effects of dioscin. This study provides further pharmacological groundwork for developing dioscin as an anti-melanoma agent.
Collapse
Affiliation(s)
- Yu-Xi Liu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Bo-Wen Xu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Xiao-Di Niu
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Ying-Jie Chen
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Xiu-Qiong Fu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Xiao-Qi Wang
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Cheng-Le Yin
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Ji-Yao Chou
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Jun-Kui Li
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Jia-Ying Wu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Jing-Xuan Bai
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Ying Wu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Sze-Man Li
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Zhi-Ling Yu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China; Research and Development Centre for Natural Health Products, HKBU Institute for Research and Continuing Education, Shenzhen, China.
| |
Collapse
|
193
|
Ismail AA, Shaker BT, Bajou K. The Plasminogen-Activator Plasmin System in Physiological and Pathophysiological Angiogenesis. Int J Mol Sci 2021; 23:ijms23010337. [PMID: 35008762 PMCID: PMC8745544 DOI: 10.3390/ijms23010337] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is a process associated with the migration and proliferation of endothelial cells (EC) to form new blood vessels. It is involved in various physiological and pathophysiological conditions and is controlled by a wide range of proangiogenic and antiangiogenic molecules. The plasminogen activator–plasmin system plays a major role in the extracellular matrix remodeling process necessary for angiogenesis. Urokinase/tissue-type plasminogen activators (uPA/tPA) convert plasminogen into the active enzyme plasmin, which in turn activates matrix metalloproteinases and degrades the extracellular matrix releasing growth factors and proangiogenic molecules such as the vascular endothelial growth factor (VEGF-A). The plasminogen activator inhibitor-1 (PAI-1) is the main inhibitor of uPA and tPA, thereby an inhibitor of pericellular proteolysis and intravascular fibrinolysis, respectively. Paradoxically, PAI-1, which is expressed by EC during angiogenesis, is elevated in several cancers and is found to promote angiogenesis by regulating plasmin-mediated proteolysis and by promoting cellular migration through vitronectin. The urokinase-type plasminogen activator receptor (uPAR) also induces EC cellular migration during angiogenesis via interacting with signaling partners. Understanding the molecular functions of the plasminogen activator plasmin system and targeting angiogenesis via blocking serine proteases or their interactions with other molecules is one of the major therapeutic strategies scientists have been attracted to in controlling tumor growth and other pathological conditions characterized by neovascularization.
Collapse
Affiliation(s)
- Asmaa Anwar Ismail
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (A.A.I.); (B.T.S.)
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Baraah Tariq Shaker
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (A.A.I.); (B.T.S.)
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Khalid Bajou
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (A.A.I.); (B.T.S.)
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence:
| |
Collapse
|
194
|
Golhani V, Ray SK, Mukherjee S. Role of MicroRNAs and Long Non-Coding RNAs in Regulating Angiogenesis in Human Breast Cancer- A Molecular Medicine Perspective. Curr Mol Med 2021; 22:882-893. [PMID: 34923940 DOI: 10.2174/1566524022666211217114527] [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: 04/07/2021] [Revised: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022]
Abstract
MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are proficient in regulating gene expression post-transcriptionally. Considering the recent trend in exploiting non-coding RNAs (ncRNAs) as cancer therapeutics, the potential use of miRNAs and lncRNAs as biomarkers and novel therapeutic agents against angiogenesis is an important scientific aspect. An estimated 70% of the genome is actively transcribed, only 2% of which codes for known protein-coding genes. Long noncoding RNAs (lncRNAs) are a large and diverse class of RNAs > 200 nucleotides in length, and not translated into protein, and are of utmost importance and it governs the expression of genes in a temporal, spatial, and cell context-dependent manner. Angiogenesis is an essential process for organ morphogenesis and growth during development, and it is relevant during the repair of wounded tissue in adults. It is coordinated by an equilibrium of pro-and anti-angiogenic factors; nevertheless, when affected, it promotes several diseases, including breast cancer. Signaling pathways involved here are tightly controlled systems that regulate the appropriate timing of gene expression required for the differentiation of cells down a particular lineage essential for proper tissue development. Lately, scientific reports are indicating that ncRNAs, such as miRNAs, and lncRNAs, play critical roles in angiogenesis related to breast cancer. The specific roles of various miRNAs and lncRNAs in regulating angiogenesis in breast cancer, with particular focus on the downstream targets and signaling pathways regulated by these ncRNAs with molecular medicine perspective, are highlighted in this write-up.
Collapse
Affiliation(s)
- Vandana Golhani
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya Pradesh-462020, India
| | | | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya Pradesh-462020, India
| |
Collapse
|
195
|
Mauro A, Berardinelli P, Russo V, Bernabò N, Martelli A, Nardinocchi D, Di Giacinto O, Turriani M, Barboni B. Effects of P 4 Antagonist RU486 on VEGF and Its Receptors' Signaling during the In Vivo Transition from the Preovulatory to Periovulatory Phase of Ovarian Follicles. Int J Mol Sci 2021; 22:13520. [PMID: 34948315 PMCID: PMC8706603 DOI: 10.3390/ijms222413520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
The development of an adequate blood vessel network is crucial for the accomplishment of ovarian follicle growth and ovulation, which is necessary to support the proliferative and endocrine functions of the follicular cells. Although the Vascular Endothelial Growth Factor (VEGF) through gonadotropins guides ovarian angiogenesis, the role exerted by the switch on of Progesterone (P4) during the periovulatory phase remains to be clarified. The present research aimed to investigate in vivo VEGF-mediated mechanisms by inducing the development of periovulatory follicles using a pharmacologically validated synchronization treatment carried out in presence or absence of P4 receptor antagonist RU486. Spatio-temporal expression profiles of VEGF, FLT1, and FLK1 receptors and the two major MAPK/ERKs and PI3K/AKT downstream pathways were analyzed on granulosa and on theca compartment. For the first time, the results demonstrated that in vivo administration of P4 antagonist RU486 inhibits follicular VEGF receptors' signaling mainly acting on the theca layer by downregulating the activation of ERKs and AKTs. Under the effect of RU486, periovulatory follicles' microarchitecture did not move towards the periovulatory stage. The present evidence provides new insights on P4 in vivo biological effects in driving vascular and tissue remodeling during the periovulatory phase.
Collapse
Affiliation(s)
- Annunziata Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.); (N.B.); (A.M.); (D.N.); (O.D.G.); (M.T.); (B.B.)
| | - Paolo Berardinelli
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.); (N.B.); (A.M.); (D.N.); (O.D.G.); (M.T.); (B.B.)
| | - Valentina Russo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.); (N.B.); (A.M.); (D.N.); (O.D.G.); (M.T.); (B.B.)
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.); (N.B.); (A.M.); (D.N.); (O.D.G.); (M.T.); (B.B.)
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council, A. Buzzati-Traverso Campus, Via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Alessandra Martelli
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.); (N.B.); (A.M.); (D.N.); (O.D.G.); (M.T.); (B.B.)
| | - Delia Nardinocchi
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.); (N.B.); (A.M.); (D.N.); (O.D.G.); (M.T.); (B.B.)
| | - Oriana Di Giacinto
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.); (N.B.); (A.M.); (D.N.); (O.D.G.); (M.T.); (B.B.)
| | - Maura Turriani
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.); (N.B.); (A.M.); (D.N.); (O.D.G.); (M.T.); (B.B.)
| | - Barbara Barboni
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.); (N.B.); (A.M.); (D.N.); (O.D.G.); (M.T.); (B.B.)
| |
Collapse
|
196
|
Imidazo[1,2-b]pyridazine as privileged scaffold in medicinal chemistry: An extensive review. Eur J Med Chem 2021; 226:113867. [PMID: 34607244 DOI: 10.1016/j.ejmech.2021.113867] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/09/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023]
Abstract
Imidazo[1,2-b]pyridazine scaffold represents an important class of heterocyclic nucleus which provides various bioactives molecules. Among them, the successful kinase inhibitor ponatinib led to a resurgence of interest in exploring new imidazo[1,2-b]pyridazine-containing derivatives for their putative therapeutic applications in medicine. This present review intends to provide a state-of-the-art of this framework in medicinal chemistry from 1966 to nowadays, unveiling different aspects of its structure-activity relationships (SAR). This extensive literature surveil may guide medicinal chemists for the quest of novel imidazo[1,2-b]pyridazine compounds with enhanced pharmacokinetics profile and efficiency.
Collapse
|
197
|
Wu XY, Zhu YM, Qi Y, Xu WW, Jing-Zhai. Erythropoietin, as a biological macromolecule in modification of tissue engineered constructs: A review. Int J Biol Macromol 2021; 193:2332-2342. [PMID: 34793816 DOI: 10.1016/j.ijbiomac.2021.11.065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/08/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022]
Abstract
In recent years, tissue engineering has emerged as a promising approach to address limitations of organ transplantation. The ultimate goal of tissue engineering is to provide scaffolds that closely mimic the physicochemical and biological cues of native tissues' extracellular matrix. In this endeavor, new generation of scaffolds have been designed that utilize the incorporation of signaling molecules in order to improve cell recruitment, enhance angiogenesis, exert healing activities, and increase the engraftment of the scaffolds. Among different signaling molecules, the role of erythropoietin (EPO) in regenerative medicine is increasingly being appreciated. It is a biological macromolecule which can prevent programed cell death, modulate inflammation, induce cell proliferation, and provide tissue protection in different disease models. In this review, we have outlined and critically analyzed different techniques of scaffolds' modification with EPO or EPO-loaded nanoparticles. We have also explored different strategies for the incorporation of EPO into scaffolds. Non-hematopoietic functions of EPO have also been discussed. Finalizing with detailed discussion surrounding the applications, challenges, and future perspectives of EPO-modified scaffolds in regenerative medicine.
Collapse
Affiliation(s)
- Xiao-Yu Wu
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Yi-Miao Zhu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, China
| | - Yang Qi
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, China
| | - Wen-Wen Xu
- Department of Gynaecology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China.
| | - Jing-Zhai
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
198
|
Rabelo ACS, Borghesi J, Noratto GD. The role of dietary polyphenols in osteosarcoma: A possible clue about the molecular mechanisms involved in a process that is just in its infancy. J Food Biochem 2021; 46:e14026. [PMID: 34873724 DOI: 10.1111/jfbc.14026] [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: 08/16/2021] [Revised: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022]
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor mainly affecting children, teenagers and young adults, being associated with early metastasis and poor prognosis. The beneficial effects of polyphenols have been investigated in different areas, including their potential to fight OS. Polyphenols are believed to reduce morbidity and/or slow down the development of cancer. This review aimed to assess the effect of polyphenols in OS and investigate their molecular mechanisms. It was observed that the broad spectrum of health-promoting properties of plant polyphenols in OS occurs mainly due to modulation of reactive oxygen species, anti-inflammatory activity, anti-angiogenesis, apoptosis inducer, inhibition of invasion and metastasis. However, it is worth mentioning that although the promising effects of polyphenols in the fight against OS, most of the studies have been performed using in vitro and in vivo animal models. Therefore, studies in humans are needed to validate the effectiveness of polyphenols in OS treatment. PRACTICAL APPLICATIONS: Polyphenols are widely used for various diseases, however, until now, their real role in the treatment of osteosarcoma remains unknown. This review provides a broad spectrum of research conducted with polyphenols and their potential as adjuvant therapy in the treatment of osteosarcoma. However, prior to their clinical application for osteosarcoma treatment, there is a need to isolate and identify specific polyphenolic compounds with high antitumor activity, increase their oral bioavailability, and to investigate their interactions with chemotherapeutic drugs being used in clinical practice.
Collapse
Affiliation(s)
- Ana Carolina Silveira Rabelo
- Department of Food and Experimental Nutrition, Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Jéssica Borghesi
- Department of Anatomy, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo, Brazil
| | - Giuliana D Noratto
- Departament of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
199
|
Shi A, Wang T, Jia M, Dong L, Shi H. Effects of SDF-1/CXCR7 on the Migration, Invasion and Epithelial-Mesenchymal Transition of Gastric Cancer Cells. Front Genet 2021; 12:760048. [PMID: 34858476 PMCID: PMC8630678 DOI: 10.3389/fgene.2021.760048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
We found that SDF-1/CXCR7 axis played an important role in the growth and proliferation of gastric cancer in the previous studies. The objectives of this study were to explore the effects of SDF-1/CXCR7 on the metastatic ability of gastric cancer cells and the possible mechanisms. CXCR7 expression in SGC-7901 gastric cancer cells was stably knocked down via lentiviral vectors. The cell migration and invasion abilities were detected by transwell migration and invasion assays. The expressions of matrix metalloproteinase 2 (MMP-2), MMP-9, vascular endothelial growth factor (VEGF), epithelial-mesenchymal transition (EMT) markers and Akt phosphorylation were detected with real-time PCR and/or western blot. We found that SDF-1 markedly enhanced the migration and invasion abilities of SGC-7901 gastric cancer cells; CXCR7 knockdown inhibited these effects. SDF-1/CXCR7 increased the expressions of MMP-2, MMP-9 and VEGF. SDF-1/CXCR7 also downregulated E-cadherin expression but upregulated N-cadherin, vimentin and Snail expressions, suggesting that SDF-1/CXCR7 could promote the development of EMT in gastric cancer cells. Furthermore, SDF-1/CXCR7 could promote Akt phosphorylation. Our results indicated that SDF-1/CXCR7 enhanced the migration, invasion and EMT of gastric cancer cells and thus CXCR7 supression may be a strategy for inhibiting gastric cancer metastasis.
Collapse
Affiliation(s)
- Ameng Shi
- Department of Ultrasound, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ting Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Miao Jia
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lei Dong
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haitao Shi
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
200
|
Gu Y, Pais G, Becker V, Körbel C, Ampofo E, Ebert E, Hohneck J, Ludwig N, Meese E, Bohle RM, Zhao Y, Menger MD, Laschke MW. Suppression of endothelial miR-22 mediates non-small cell lung cancer cell-induced angiogenesis. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:849-864. [PMID: 34729252 PMCID: PMC8536510 DOI: 10.1016/j.omtn.2021.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/06/2021] [Accepted: 10/01/2021] [Indexed: 11/16/2022]
Abstract
MicroRNAs (miRNAs) expressed in endothelial cells (ECs) are powerful regulators of angiogenesis, which is essential for tumor growth and metastasis. Here, we demonstrated that miR-22 is preferentially and highly expressed in ECs, while its endothelial level is significantly downregulated in human non-small cell lung cancer (NSCLC) tissues when compared to matched nontumor lung tissues. This reduction of endothelial miR-22 is possibly induced by NSCLC cell-secreted interleukin-1β and subsequently activated transcription factor nuclear factor-κB. Endothelial miR-22 functions as a potent angiogenesis inhibitor that inhibits all of the key angiogenic activities of ECs and consequently NSCLC growth through directly targeting sirtuin 1 and fibroblast growth factor receptor 1 in ECs, leading to inactivation of AKT/mammalian target of rapamycin signaling. These findings provide insight into the molecular mechanisms of NSCLC angiogenesis and indicate that endothelial miR-22 represents a potential target for the future antiangiogenic treatment of NSCLC.
Collapse
Affiliation(s)
- Yuan Gu
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Gianni Pais
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Vivien Becker
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Christina Körbel
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Elke Ebert
- Institute of Pathology, Saarland University, 66421 Homburg/Saar, Germany
| | - Johannes Hohneck
- Institute of Pathology, Saarland University, 66421 Homburg/Saar, Germany
| | - Nicole Ludwig
- Institute of Human Genetics, Saarland University, 66421 Homburg/Saar, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, 66421 Homburg/Saar, Germany
| | - Rainer M. Bohle
- Institute of Pathology, Saarland University, 66421 Homburg/Saar, Germany
| | - Yingjun Zhao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Michael D. Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Matthias W. Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| |
Collapse
|