1
|
Das K, Schulte M, Gerhart M, Bayoumi H, Paulson D, Fink DM, Parrish C, Willand-Charnley R. Early in vitro results indicate that de-O-acetylated sialic acids increase Selectin binding in cancers. Front Oncol 2024; 14:1443303. [PMID: 39717751 PMCID: PMC11663943 DOI: 10.3389/fonc.2024.1443303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/11/2024] [Indexed: 12/25/2024] Open
Abstract
Cancers utilize a simple glycan, Sialic Acid, to engage in metastatic processes via the Sialic acid (Sia) -Selectin pathway. Selectins recognize and bind to sialylated substrates, resulting in adhesion, migration, and extravasation, however, how deviations from the canonical form of Sia regulate binding to Selectin receptors (E, L, and P) on hemopoietic cells resulting in these metastatic processes, remained a gap in knowledge. De-O-acetylated Sias has been recently shown to be an integral substrate to the binding of sialic acid binding proteins. The two proteins responsible for regulating the acetyl functional group on Sia's C6 tail, are Sialic acid acetylesterase (SIAE) and Sialic acid O acetyltransferase (CASD1). To elucidate the contribution of functional group alterations on Sia, 9-O and 7,9-O-acetylation of Sia was modulated via the use of CRISRP-Cas9 gene editing and with Sialyl Glycan Recognition Probes, to produce either O-acetylated-Sia or de-O-acetylated- Sia, respectively. In vitro experiments revealed that increased cell surface expression of de-O-acetylated- Sia resulted in an increase in Selectin binding, enhanced cell proliferation, and increased migration capabilities both in lung and colon cancer. These results delineate for the first time the mechanistic contribution of de-O-acetylated-Sia to Selectin binding, reinforcing the importance of elucidating functional group alterations on Sia and their contribution. Without accurate identification of which functionalized form of Sia is being utilized to bind to sialic acid binding proteins, we cannot accurately produce glycan therapeutics with the correct specificity and reactivity, thus this work contributes an integral component in the development of promising therapeutic avenues, for example in the realm of enzyme antibody drug conjugates.
Collapse
Affiliation(s)
- Kakali Das
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Megan Schulte
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Megan Gerhart
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Hala Bayoumi
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Delayna Paulson
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Darci M. Fink
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Colin Parrish
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Rachel Willand-Charnley
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
2
|
Kheiri S, Yakavets I, Cruickshank J, Ahmadi F, Berman HK, Cescon DW, Young EWK, Kumacheva E. Microfluidic Platform for Generating and Releasing Patient-Derived Cancer Organoids with Diverse Shapes: Insight into Shape-Dependent Tumor Growth. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410547. [PMID: 39276011 DOI: 10.1002/adma.202410547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/15/2024] [Indexed: 09/16/2024]
Abstract
Multicellular spheroids and patient-derived organoids find many applications in fundamental research, drug discovery, and regenerative medicine. Advances in the understanding and recapitulation of organ functionality and disease development require the generation of complex organoid models, including organoids with diverse morphologies. Microfluidics-based cell culture platforms enable time-efficient confined organoid generation. However, the ability to form organoids with different shapes with a subsequent transfer from microfluidic devices to unconstrained environments for studies of morphology-dependent organoid growth is yet to be demonstrated. Here, a microfluidic platform is introduced that enables high-fidelity formation and addressable release of breast cancer organoids with diverse shapes. Using this platform, the impact of organoid morphology on their growth in unconstrained biomimetic hydrogel is explored. It is shown that proliferative cancer cells tend to localize in high positive curvature organoid regions, causing their faster growth, while the overall growth pattern of organoids with diverse shapes tends to reduce interfacial tension at the organoid-hydrogel interface. In addition to the formation of organoids with diverse morphologies, this platform can be integrated into multi-tissue micro-physiological systems.
Collapse
Affiliation(s)
- Sina Kheiri
- Department of Mechanical & Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, M5S 3G8, Canada
| | - Ilya Yakavets
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada
| | - Jennifer Cruickshank
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, M5G 2C1, Canada
| | - Fatemeh Ahmadi
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada
| | - Hal K Berman
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, M5G 2C1, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, M5G 2C1, Canada
- Department of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Edmond W K Young
- Department of Mechanical & Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
| | - Eugenia Kumacheva
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
| |
Collapse
|
3
|
Ma K, Wang S, Ma Y, Zeng L, Xu K, Mu N, Lai Y, Shi Y, Yang C, Chen B, Quan Y, Li L, Lu Y, Yang Y, Liu Y, Hu R, Wang X, Chen Y, Bian X, Feng H, Li F, Chen T. Increased oxygen stimulation promotes chemoresistance and phenotype shifting through PLCB1 in gliomas. Drug Resist Updat 2024; 76:101113. [PMID: 39053384 DOI: 10.1016/j.drup.2024.101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/24/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
Gliomas, the most common CNS (central nerve system) tumors, face poor survival due to severe chemoresistance exacerbated by hypoxia. However, studies on whether altered hypoxic conditions benefit for chemo-sensitivity and how gliomas react to increased oxygen stimulation are limited. In this study, we demonstrated that increased oxygen stimulation promotes glioma growth and chemoresistance. Mechanically, increased oxygen stimulation upregulates miR-1290 levels. miR-1290, in turn, downregulates PLCB1, while PLCB1 facilitates the proteasomal degradation of β-catenin and active-β-catenin by increasing the proportion of ubiquitinated β-catenin in a destruction complex-independent mechanism. This process inhibits PLCB1 expression, leads to the accumulation of active-β-catenin, boosting Wnt signaling through an independent mechanism and ultimately promoting chemoresistance in glioma cells. Pharmacological inhibition of Wnt by WNT974 could partially inhibit glioma volume growth and prolong the shortened survival caused by increased oxygen stimulation in a glioma-bearing mouse model. Moreover, PLCB1, a key molecule regulated by increased oxygen stimulation, shows promising predictive power in survival analysis and has great potential to be a biomarker for grading and prognosis in glioma patients. These results provide preliminary insights into clinical scenarios associated with altered hypoxic conditions in gliomas, and introduce a novel perspective on the role of the hypoxic microenvironment in glioma progression. Furthermore, the outcomes reveal the potential risks of utilizing hyperbaric oxygen treatment (HBOT) in glioma patients, particularly when considering HBOT as a standalone option to ameliorate neuro-dysfunctions or when combining HBOT with a single chemotherapy agent without radiotherapy.
Collapse
Affiliation(s)
- Kang Ma
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shi Wang
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yingjie Ma
- Medical Data Science Academy, Chongqing Medical University, Chongqing, China
| | - Lan Zeng
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Kai Xu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ning Mu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Lai
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yaning Shi
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chuanyan Yang
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Beike Chen
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yulian Quan
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Li
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yongling Lu
- Medical Research Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yang Yang
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yan Liu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Rong Hu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoming Wang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yujie Chen
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Hua Feng
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Fei Li
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Tunan Chen
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
4
|
Stribbling SM, Beach C, Ryan AJ. Orthotopic and metastatic tumour models in preclinical cancer research. Pharmacol Ther 2024; 257:108631. [PMID: 38467308 DOI: 10.1016/j.pharmthera.2024.108631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
Mouse models of disease play a pivotal role at all stages of cancer drug development. Cell-line derived subcutaneous tumour models are predominant in early drug discovery, but there is growing recognition of the importance of the more complex orthotopic and metastatic tumour models for understanding both target biology in the correct tissue context, and the impact of the tumour microenvironment and the immune system in responses to treatment. The aim of this review is to highlight the value that orthotopic and metastatic models bring to the study of tumour biology and drug development while pointing out those models that are most likely to be encountered in the literature. Important developments in orthotopic models, such as the increasing use of early passage patient material (PDXs, organoids) and humanised mouse models are discussed, as these approaches have the potential to increase the predictive value of preclinical studies, and ultimately improve the success rate of anticancer drugs in clinical trials.
Collapse
Affiliation(s)
- Stephen M Stribbling
- Department of Chemistry, University College London, Gower Street, London WC1E 6BT, UK.
| | - Callum Beach
- Department of Oncology, University of Oxford, ORCRB, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Anderson J Ryan
- Department of Oncology, University of Oxford, ORCRB, Roosevelt Drive, Oxford OX3 7DQ, UK; Fast Biopharma, Aston Rowant, Oxfordshire, OX49 5SW, UK.
| |
Collapse
|
5
|
Huang X, Tang Q, Liu S, Li C, Li Y, Sun Y, Ding X, Xia L, Hu S. Discovery of an antitumor compound from xenorhabdus stockiae HN_xs01. World J Microbiol Biotechnol 2024; 40:101. [PMID: 38366186 DOI: 10.1007/s11274-024-03915-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024]
Abstract
Xenorhabdus, known for its symbiotic relationship with Entomopathogenic nematodes (EPNs), belongs to the Enterobacteriaceae family. This dual-host symbiotic nematode exhibits pathogenic traits, rendering it a promising biocontrol agent against insects. Our prior investigations revealed that Xenorhabdus stockiae HN_xs01, isolated in our laboratory, demonstrates exceptional potential in halting bacterial growth and displaying anti-tumor activity. Subsequently, we separated and purified the supernatant of the HN_xs01 strain and obtained a new compound with significant inhibitory activity on tumor cells, which we named XNAE. Through LC-MS analysis, the mass-to-nucleus ratio of XNAE was determined to be 254.24. Our findings indicated that XNAE exerts a time- and dose-dependent inhibition on B16 and HeLa cells. After 24 h, its IC50 for B16 and HeLa cells was 30.178 µg/mL and 33.015 µg/mL, respectively. Electron microscopy revealed conspicuous damage to subcellular structures, notably mitochondria and the cytoskeleton, resulting in a notable reduction in cell numbers among treated tumor cells. Interestingly, while XNAE exerted a more pronounced inhibitory effect on B16 cells compared to HeLa cells, it showed no discernible impact on HUVEC cells. Treatment of B16 cells with XNAE induced early apoptosis and led to cell cycle arrest in the G2 phase, as evidenced by flow cytometry analysis. The impressive capability of X. stockiae HN_xs01 in synthesizing bioactive secondary metabolites promises to significantly expand the reservoir of natural products. Further exploration to identify the bioactivity of these compounds holds the potential to shed light on their roles in bacteria-host interaction. Overall, these outcomes underscore the promising potential of XNAE as a bioactive compound for tumor treatment.
Collapse
Affiliation(s)
- Xiyin Huang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No.36 Lushan Street, Changsha, 410081, China
| | - Qiong Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No.36 Lushan Street, Changsha, 410081, China
| | - Siqin Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No.36 Lushan Street, Changsha, 410081, China
| | - Chen Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No.36 Lushan Street, Changsha, 410081, China
| | - Yaoguang Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No.36 Lushan Street, Changsha, 410081, China
| | - Yunjun Sun
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No.36 Lushan Street, Changsha, 410081, China
| | - Xuezhi Ding
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No.36 Lushan Street, Changsha, 410081, China
| | - Liqiu Xia
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No.36 Lushan Street, Changsha, 410081, China
| | - Shengbiao Hu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No.36 Lushan Street, Changsha, 410081, China.
| |
Collapse
|
6
|
Marchello R, Colombi A, Preziosi L, Giverso C. A non local model for cell migration in response to mechanical stimuli. Math Biosci 2024; 368:109124. [PMID: 38072125 DOI: 10.1016/j.mbs.2023.109124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 11/17/2023] [Accepted: 12/05/2023] [Indexed: 12/22/2023]
Abstract
Cell migration is one of the most studied phenomena in biology since it plays a fundamental role in many physiological and pathological processes such as morphogenesis, wound healing and tumorigenesis. In recent years, researchers have performed experiments showing that cells can migrate in response to mechanical stimuli of the substrate they adhere to. Motion towards regions of the substrate with higher stiffness is called durotaxis, while motion guided by the stress or the deformation of the substrate itself is called tensotaxis. Unlike chemotaxis (i.e. the motion in response to a chemical stimulus), these migratory processes are not yet fully understood from a biological point of view. In this respect, we present a mathematical model of single-cell migration in response to mechanical stimuli, in order to simulate these two processes. Specifically, the cell moves by changing its direction of polarization and its motility according to material properties of the substrate (e.g., stiffness) or in response to proper scalar measures of the substrate strain or stress. The equations of motion of the cell are non-local integro-differential equations, with the addition of a stochastic term to account for random Brownian motion. The mechanical stimulus to be integrated in the equations of motion is defined according to experimental measurements found in literature, in the case of durotaxis. Conversely, in the case of tensotaxis, substrate strain and stress are given by the solution of the mechanical problem, assuming that the extracellular matrix behaves as a hyperelastic Yeoh's solid. In both cases, the proposed model is validated through numerical simulations that qualitatively reproduce different experimental scenarios.
Collapse
Affiliation(s)
- Roberto Marchello
- Mathematics Area, SISSA (International School for Advanced Studies), Via Bonomea 265, Trieste, 34136, Italy
| | - Annachiara Colombi
- Department of Mathematical Sciences G. L. Lagrange, Politecnico di Torino, C.so Duca degli Abruzzi 24, Torino, 10129, Italy
| | - Luigi Preziosi
- Department of Mathematical Sciences G. L. Lagrange, Politecnico di Torino, C.so Duca degli Abruzzi 24, Torino, 10129, Italy
| | - Chiara Giverso
- Department of Mathematical Sciences G. L. Lagrange, Politecnico di Torino, C.so Duca degli Abruzzi 24, Torino, 10129, Italy.
| |
Collapse
|
7
|
Umar H, Aliyu MR, Usman AG, Ghali UM, Abba SI, Ozsahin DU. Prediction of cell migration potential on human breast cancer cells treated with Albizia lebbeck ethanolic extract using extreme machine learning. Sci Rep 2023; 13:22242. [PMID: 38097683 PMCID: PMC10721884 DOI: 10.1038/s41598-023-49363-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023] Open
Abstract
Cancer is one of the major causes of death in the modern world, and the incidence varies considerably based on race, ethnicity, and region. Novel cancer treatments, such as surgery and immunotherapy, are ineffective and expensive. In this situation, ion channels responsible for cell migration have appeared to be the most promising targets for cancer treatment. This research presents findings on the organic compounds present in Albizia lebbeck ethanolic extracts (ALEE), as well as their impact on the anti-migratory, anti-proliferative and cytotoxic potentials on MDA-MB 231 and MCF-7 human breast cancer cell lines. In addition, artificial intelligence (AI) based models, multilayer perceptron (MLP), extreme gradient boosting (XGB), and extreme learning machine (ELM) were performed to predict in vitro cancer cell migration on both cell lines, based on our experimental data. The organic compounds composition of the ALEE was studied using gas chromatography-mass spectrometry (GC-MS) analysis. Cytotoxicity, anti-proliferations, and anti-migratory activity of the extract using Tryphan Blue, MTT, and Wound Heal assay, respectively. Among the various concentrations (2.5-200 μg/mL) of the ALEE that were used in our study, 2.5-10 μg/mL revealed anti-migratory potential with increased concentrations, and they did not show any effect on the proliferation of the cells (P < 0.05; n ≥ 3). Furthermore, the three data-driven models, Multi-layer perceptron (MLP), Extreme gradient boosting (XGB), and Extreme learning machine (ELM), predict the potential migration ability of the extract on the treated cells based on our experimental data. Overall, the concentrations of the plant extract that do not affect the proliferation of the type cells used demonstrated promising effects in reducing cell migration. XGB outperformed the MLP and ELM models and increased their performance efficiency by up to 3% and 1% for MCF and 1% and 2% for MDA-MB231, respectively, in the testing phase.
Collapse
Affiliation(s)
- Huzaifa Umar
- Near East University, Operational Research Centre in Healthcare, TRNC Mersin 10, 99138, Nicosia, Turkey.
| | - Maryam Rabiu Aliyu
- Department of Energy System Engineering, Cyprus International University, Northern Cyprus via Mersin 10, 99258, Nicosia, Turkey
| | - Abdullahi Garba Usman
- Near East University, Operational Research Centre in Healthcare, TRNC Mersin 10, 99138, Nicosia, Turkey
- Department of Analytical Chemistry, Faculty of Pharmacy, Near East University, TRNC, Mersin 10, 99138, Nicosia, Turkey
| | - Umar Muhammad Ghali
- Department of Chemistry, Faculty of Natural and Applied Sciences, Firat University, Merkezi, 23199, Elazig, Turkey
| | - Sani Isah Abba
- Interdisciplinary Research Centre for Membranes and Water Security, King Fahd University of Petroleum and Minerals, 31261, Dhahran, Saudi Arabia
| | - Dilber Uzun Ozsahin
- Department of Medical Diagnostic Imaging, College of Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
8
|
Francelino AL, Pereira ÉR, Pinheiro LCL, Soares AC, Mendonça AC, Fuganti PE, Frantine-Silva W, de Oliveira KB, Serpeloni JM, Guembarovski RL. Allelic variants and immunostaining profile in CXCL12/CXCR4 axis: An investigation of association with prognosis in prostate cancer. Pathol Res Pract 2023; 250:154806. [PMID: 37738928 DOI: 10.1016/j.prp.2023.154806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/24/2023]
Abstract
Prostate cancer (PCa) is the malignant neoplasm that most commonly affects men and is an important cause of death. It can be detected by changes in serum levels of Prostate Specific Antigen (PSA) and digital rectal examination, but often symptoms do not appear until advanced stages and metastases. The C-X-C Motif Chemokine Ligand 12/C-X-C Motif Chemokine Receptor 4 (CXCL12/CXCR4) axis acts in cell migration and may be involved in the metastatic process. In this context, the aim of this study was to evaluate the allelic variants rs1801157 (CXCL12) and rs2228014 (CXCR4) and the immunostaining of CXCR4 protein as candidates for prognostic markers in PCa. Samples (n = 60) were divided according to prognostic parameters (with and without metastasis at diagnosis) in tree groups: better prognosis, worse prognosis with metastasis at diagnosis and worse prognosis without metastasis at diagnosis, and immunostaining was evaluated by indirect immunohistochemistry, considering tumoral and adjacent tissues from the same patient (n = 120). A significant association was found between the C allele of rs2228014 (CXCR4) and the extraprostatic extension. For CXCR4 immunostaining a weak labeling and a cytoplasmic localization predominated, as well as a significant difference between malignant versus adjacent tissue, with higher protein expression in the malignant tissue. A significant association was found between CXCR4 tumor immunostaining with TNM staging (T2b-T2c) and PSA level (> 20 ng/mL). None of the allelic variants affected CXCR4 immunostaining. Prognostic groups did not differ in allelic variant frequency or immunostaining profile. Findings suggest that CXCR4 receptor may be one of the ways to worsen the prognosis of prostatic cancer.
Collapse
Affiliation(s)
- Amanda Letícia Francelino
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Érica Romão Pereira
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Laís Capelasso Lucas Pinheiro
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Adrielli Caroline Soares
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Ana Carla Mendonça
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | | | - Wilson Frantine-Silva
- Laboratory of Studies and Applications of DNA Polymorphism, Department of Immunology, Parasitology and General Pathology, CCB, Londrina State University, Londrina, PR, Brazil
| | - Karen Brajão de Oliveira
- Laboratory of Molecular Genetics and Immunology, Department of Pathological Sciences, Londrina State University, Londrina, PR, Brazil; Laboratory of Studies and Applications of DNA Polymorphism, Department of Immunology, Parasitology and General Pathology, CCB, Londrina State University, Londrina, PR, Brazil
| | - Juliana Mara Serpeloni
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Roberta Losi Guembarovski
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil.
| |
Collapse
|
9
|
Bloomfield M, Cimini D. The fate of extra centrosomes in newly formed tetraploid cells: should I stay, or should I go? Front Cell Dev Biol 2023; 11:1210983. [PMID: 37576603 PMCID: PMC10413984 DOI: 10.3389/fcell.2023.1210983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
An increase in centrosome number is commonly observed in cancer cells, but the role centrosome amplification plays along with how and when it occurs during cancer development is unclear. One mechanism for generating cancer cells with extra centrosomes is whole genome doubling (WGD), an event that occurs in over 30% of human cancers and is associated with poor survival. Newly formed tetraploid cells can acquire extra centrosomes during WGD, and a generally accepted model proposes that centrosome amplification in tetraploid cells promotes cancer progression by generating aneuploidy and chromosomal instability. Recent findings, however, indicate that newly formed tetraploid cells in vitro lose their extra centrosomes to prevent multipolar cell divisions. Rather than persistent centrosome amplification, this evidence raises the possibility that it may be advantageous for tetraploid cells to initially restore centrosome number homeostasis and for a fraction of the population to reacquire additional centrosomes in the later stages of cancer evolution. In this review, we explore the different evolutionary paths available to newly formed tetraploid cells, their effects on centrosome and chromosome number distribution in daughter cells, and their probabilities of long-term survival. We then discuss the mechanisms that may alter centrosome and chromosome numbers in tetraploid cells and their relevance to cancer progression following WGD.
Collapse
Affiliation(s)
- Mathew Bloomfield
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| | - Daniela Cimini
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
10
|
Quadri M, Tiso N, Musmeci F, Morasso MI, Brooks SR, Bonetti LR, Panini R, Lotti R, Marconi A, Pincelli C, Palazzo E. CD271 activation prevents low to high-risk progression of cutaneous squamous cell carcinoma and improves therapy outcomes. J Exp Clin Cancer Res 2023; 42:167. [PMID: 37443031 DOI: 10.1186/s13046-023-02737-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (cSCC) is the second most prevalent form of skin cancer, showing a rapid increasing incidence worldwide. Although most cSCC can be cured by surgery, a sizeable number of cases are diagnosed at advanced stages, with local invasion and distant metastatic lesions. In the skin, neurotrophins (NTs) and their receptors (CD271 and Trk) form a complex network regulating epidermal homeostasis. Recently, several works suggested a significant implication of NT receptors in cancer. However, CD271 functions in epithelial tumors are controversial and its precise role in cSCC is still to be defined. METHODS Spheroids from cSCC patients with low-risk (In situ or Well-Differentiated cSCC) or high-risk tumors (Moderately/Poorly Differentiated cSCC), were established to explore histological features, proliferation, invasion abilities, and molecular pathways modulated in response to CD271 overexpression or activation in vitro. The effect of CD271 activities on the response to therapeutics was also investigated. The impact on the metastatic process and inflammation was explored in vivo and in vitro, by using zebrafish xenograft and 2D/3D models. RESULTS Our data proved that CD271 is upregulated in Well-Differentiated tumors as compared to the more aggressive Moderately/Poorly Differentiated cSCC, both in vivo and in vitro. We demonstrated that CD271 activities reduce proliferation and malignancy marker expression in patient-derived cSCC spheroids at each tumor grade, by increasing neoplastic cell differentiation. CD271 overexpression significantly increases cSCC spheroid mass density, while it reduces their weight and diameter, and promotes a major fold-enrichment in differentiation and keratinization genes. Moreover, both CD271 overexpression and activation decrease cSCC cell invasiveness in vitro. A significant inhibition of the metastatic process by CD271 was observed in a newly established zebrafish cSCC model. We found that the recruitment of leucocytes by CD271-overexpressing cells directly correlates with tumor killing and this finding was further highlighted by monocyte infiltration in a THP-1-SCC13 3D model. Finally, CD271 activity synergizes with Trk receptor inhibition, by reducing spheroid viability, and significantly improves the outcome of photodynamic therapy (PTD) or chemotherapy in spheroids and zebrafish. CONCLUSION Our study provides evidence that CD271 could prevent the switch between low to high-risk cSCC tumors. Because CD271 contributes to maintaining active differentiative paths and favors the response to therapies, it might be a promising target for future pharmaceutical development.
Collapse
Affiliation(s)
- Marika Quadri
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy
| | - Natascia Tiso
- Laboratory of Developmental Genetics, Department of Biology, University of Padova, Padova, Italy
| | | | - Maria I Morasso
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, USA
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, USA
| | - Luca Reggiani Bonetti
- Department of Diagnostic, Clinic and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Rossana Panini
- Department of Diagnostic, Clinic and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberta Lotti
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy
| | - Alessandra Marconi
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy
| | - Carlo Pincelli
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy
| | - Elisabetta Palazzo
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy.
| |
Collapse
|
11
|
Fan X, Meng M, Li B, Chen H, Tan J, Xu K, Xiao S, Kwan HY, Liu Z, Su T. Brevilin A is a potent anti-metastatic CRC agent that targets the VEGF-IL6-STAT3 axis in the HSCs-CRC interplay. J Transl Med 2023; 21:260. [PMID: 37062842 PMCID: PMC10105967 DOI: 10.1186/s12967-023-04087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/25/2023] [Indexed: 04/18/2023] Open
Abstract
BACKGROUND More than half of the colorectal cancer (CRC) patients will develop liver metastasis that underlies the cancer mortality. In the hepatic tumor microenvironment, the interplay between CRC cells and hepatic stellate cells (HSCs), and the activation of HSCs to become carcinoma-associated fibroblasts (CAFs) will further promote the cancer development. Nevertheless, the critical signaling molecule that involved in these processes remains unknown, which hinders the development of effective therapeutic agents for the treatment of metastatic CRC (mCRC). METHODS Conditioned medium system and co-cultured system were used to examine the interplay between CRC cells and HSCs. Luminex liquid suspension chip detection and enzyme-linked immunosorbent assay were used to screen for the mediators in the conditioned medium that facilitated the CRC-HSCs interplay and HSCs-to-CAFs differentiation. Cell and animal models were used to examine whether brevilin A inhibited CRC liver metastasis via the VEGF-IL6-STAT3 axis. RESULTS In the CRC-HSCs interplay, CRC promoted HSCs-to-CAFs differentiation by releasing vascular endothelial growth factor (VEGF); and HSCs released interleukin 6 (IL6) that activated signal transducer and activator of transcription 3 (STAT3) in the CRC and hence increased the cancer metastatic potential. The functions of the VEGF-IL6-STAT3 axis in the HSCs-CRC interplay were further validated by VEGF recombinant protein and IL6 neutralizing antibody. More importantly, brevilin A, an active compound isolated from Centipeda minima (L.) A. Br. et Aschers, targeted the VEGF-IL6-STAT3 axis in the CRC-HSCs interplay, hence significantly inhibited colorectal liver metastasis and cancer growth both in vitro and in vivo. CONCLUSIONS We are the first to demonstrate brevilin A possesses potent anti-mCRC effect by targeting the VEGF-IL6-STAT3 axis in the CRC-HSCs interplay. Our findings not only support the development of brevilin A as a novel therapeutic agent for mCRC treatment, but also pave the path for the development of other VEGF-IL6-STAT3 targeting therapeutic strategies.
Collapse
Affiliation(s)
- Xueying Fan
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Mingjing Meng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Baoting Li
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Hui Chen
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jincheng Tan
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Keyang Xu
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Shilin Xiao
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hiu-Yee Kwan
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Zhongqiu Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Tao Su
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
Faggioli F, Velarde MC, Wiley CD. Cellular Senescence, a Novel Area of Investigation for Metastatic Diseases. Cells 2023; 12:cells12060860. [PMID: 36980201 PMCID: PMC10047218 DOI: 10.3390/cells12060860] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Metastasis is a systemic condition and the major challenge among cancer types, as it can lead to multiorgan vulnerability. Recently, attention has been drawn to cellular senescence, a complex stress response condition, as a factor implicated in metastatic dissemination and outgrowth. Here, we examine the current knowledge of the features required for cells to invade and colonize secondary organs and how senescent cells can contribute to this process. First, we describe the role of senescence in placentation, itself an invasive process which has been linked to higher rates of invasive cancers. Second, we describe how senescent cells can contribute to metastatic dissemination and colonization. Third, we discuss several metabolic adaptations by which senescent cells could promote cancer survival along the metastatic journey. In conclusion, we posit that targeting cellular senescence may have a potential therapeutic efficacy to limit metastasis formation.
Collapse
Affiliation(s)
- Francesca Faggioli
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
- Istituto di Ricerca Genetica e Biomedica (IRGB-CNR) uos Milan, Via Fantoli 15/16, 20090 Milan, Italy
- Correspondence: ; Tel.: +39-02-82245211
| | - Michael C. Velarde
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City PH 1101, Philippines
| | - Christopher D. Wiley
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA 02111, USA
- School of Medicine, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
13
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
14
|
Yin Z, Shi R, Xia X, Li L, Yang Y, Li S, Xu J, Xu Y, Cai X, Wang S, Liu Z, Peng T, Peng Y, Wang H, Ye M, Liu Y, Chen Z, Tan W. An Implantable Magnetic Vascular Scaffold for Circulating Tumor Cell Removal In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2207870. [PMID: 36271719 DOI: 10.1002/adma.202207870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/11/2022] [Indexed: 06/16/2023]
Abstract
An integrated trapped device (ITD) capable of removal of circulating tumor cells (CTCs) can assuage or even prevent metastasis. However, adhesion repertoires are ordinarily neglected in the design of ITDs, possibly leading to the omission of highly metastatic CTC and treatment failure. Here a vascular-like ITD with adhesive sites and wireless magnetothermal response to remove highly metastatic CTC in vivo is presented. Such a vascular-like ITD comprises circumferential well-aligned fibers and artificial adhesion repertoires and is optimized for magnetothermal integration. Continuous and repeated capture in a dynamic environment increases capture efficiency over time. Meanwhile, the heat generation of the ITD leads to the capture of CTC death owing to cell heat sensitivity. Furthermore, the constructed bioinspired ultrastructure of the ITD prevents vascular blockage and induces potential vascular regeneration. Overall, this work defines an extendable strategy for constructing adhesion repertoires against intravascular shear forces, provides a vascular-like ITD for reducing CTC counts, and is expected to alleviate the risk of cancer recurrence.
Collapse
Affiliation(s)
- Zhiwei Yin
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Rui Shi
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Xin Xia
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Ling Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yanxia Yang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Shengkai Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Jieqiong Xu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Yiting Xu
- Key Laboratory of Theoretical Organic Chemistry and Function Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, 411201, China
| | - Xinqi Cai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Shen Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Zhangkun Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Tianhuan Peng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Ying Peng
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Hua Wang
- Pediatric Research Institute, Hunan Children's Hospital, Changsha, 410007, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Zhuo Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
15
|
Bell ES, Shah P, Zuela-Sopilniak N, Kim D, Varlet AA, Morival JL, McGregor AL, Isermann P, Davidson PM, Elacqua JJ, Lakins JN, Vahdat L, Weaver VM, Smolka MB, Span PN, Lammerding J. Low lamin A levels enhance confined cell migration and metastatic capacity in breast cancer. Oncogene 2022; 41:4211-4230. [PMID: 35896617 PMCID: PMC9925375 DOI: 10.1038/s41388-022-02420-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
Abstract
Aberrations in nuclear size and shape are commonly used to identify cancerous tissue. However, it remains unclear whether the disturbed nuclear structure directly contributes to the cancer pathology or is merely a consequence of other events occurring during tumorigenesis. Here, we show that highly invasive and proliferative breast cancer cells frequently exhibit Akt-driven lower expression of the nuclear envelope proteins lamin A/C, leading to increased nuclear deformability that permits enhanced cell migration through confined environments that mimic interstitial spaces encountered during metastasis. Importantly, increasing lamin A/C expression in highly invasive breast cancer cells reflected gene expression changes characteristic of human breast tumors with higher LMNA expression, and specifically affected pathways related to cell-ECM interactions, cell metabolism, and PI3K/Akt signaling. Further supporting an important role of lamins in breast cancer metastasis, analysis of lamin levels in human breast tumors revealed a significant association between lower lamin A levels, Akt signaling, and decreased disease-free survival. These findings suggest that downregulation of lamin A/C in breast cancer cells may influence both cellular physical properties and biochemical signaling to promote metastatic progression.
Collapse
Affiliation(s)
- Emily S. Bell
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY,Current address: Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA
| | - Pragya Shah
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | | | - Dongsung Kim
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Alice-Anais Varlet
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Julien L.P. Morival
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Alexandra L. McGregor
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Philipp Isermann
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | | | - Joshua J. Elacqua
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Jonathan N. Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Linda Vahdat
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA,Helen Diller Cancer Center, Department of Bioengineering and Therapeutic Sciences, and Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA
| | - Marcus B. Smolka
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Paul N. Span
- Department of Radiation Oncology, Radiotherapy & OncoImmunology laboratory, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA. .,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
16
|
Lee SB, Hwang SJ, Son CG. CGX, a standardized herbal syrup, inhibits colon-liver metastasis by regulating the hepatic microenvironments in a splenic injection mouse model. Front Pharmacol 2022; 13:906752. [PMID: 36105183 PMCID: PMC9465806 DOI: 10.3389/fphar.2022.906752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/25/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Colon-liver metastasis is observed in approximately 50% of patients with colorectal cancer and is a critical risk factor for a low survival rate. Several clinical studies have reported that colon-liver metastasis is accelerated by pathological hepatic microenvironments such as hepatic steatosis or fibrosis. Chunggan syrup (CGX), a standardized 13-herbal mixture, has been prescribed to patients with chronic liver diseases, including fatty liver, inflammation and fibrotic change, based on preclinical and clinical evidence. Aim of the study: In the present study, we investigated anti-liver metastatic the effects of CGX in a murine colon carcinoma (MC38)-splenic injection mouse model. Materials and methods: C57BL/6N mice were administered with CGX (100, 200 or 400 mg/kg) for 14 days before or after MC38-splenic injection under normal and high-fat diet (HFD) fed conditions. Also, above experiment was repeated without MC38-splenic injection to explore underlying mechanism. Results: The number of tumor nodules and liver weight with tumors were sup-pressed by preadministration of CGX in both normal and HFD fed mice. Regarding its mechanisms, we found that CGX administration significantly activated epithelial-cadherin (E-cadherin), but decreased vascular endothelial-cadherin (VE-cadherin) in hepatic tissues under MC38-free conditions. In addition, CGX administration significantly reduced hepatic steatosis, via modulation of lipolytic and lipogenic molecules, including activated adenosine monophosphate activated protein kinase (AMPK) and peroxisome proliferator activated receptor-alpha (PPARα). Conclusion: The present data indicate that CGX exerts an anti-colon-liver metastatic property via modulation of hepatic lipid related microenvironments.
Collapse
|
17
|
Wei X, Zhang C, He S, Huang J, Huang J, Liew SS, Zeng Z, Pu K. A Dual-Locked Activatable Phototheranostic Probe for Biomarker-Regulated Photodynamic and Photothermal Cancer Therapy. Angew Chem Int Ed Engl 2022; 61:e202202966. [PMID: 35396786 DOI: 10.1002/anie.202202966] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Indexed: 12/29/2022]
Abstract
Activatable phototheranostics holds promise for precision cancer treatment owing to the "turn-on" signals and therapeutic effects. However, most activatable phototheranostic probes only possess photodynamic therapy (PDT) or photothermal therapy (PTT), which suffer from poor therapeutic efficacy due to deficient cellular oxygen and complex tumor microenvironment. We herein report a dual-locked activatable phototheranostic probe that activates near-infrared fluorescence (NIRF) signals in tumor, triggers PDT in response to a tumor-periphery biomarker, and switches from PDT to PTT upon detecting a tumor-core-hypoxia biomarker. This PDT-PTT auto-regulated probe exhibits complete tumor ablation under the photoirradiation of a single laser source by producing cytotoxic singlet oxygen at the tumor periphery and generating hyperthermia at tumor-core where is too hypoxic for PDT. This dual-locked probe represents a promising molecular design approach toward precise cancer phototheranostics.
Collapse
Affiliation(s)
- Xin Wei
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Chi Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Shasha He
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jiaguo Huang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jingsheng Huang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Si Si Liew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Ziling Zeng
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| |
Collapse
|
18
|
K Shukla S, Nguyen V, Goyal M, Gupta V. Cationically modified inhalable nintedanib niosomes: enhancing therapeutic activity against non-small-cell lung cancer. Nanomedicine (Lond) 2022; 17:935-958. [PMID: 36004583 PMCID: PMC9583758 DOI: 10.2217/nnm-2022-0045] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 07/21/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: This study was designed to develop and test nintedanib-loaded niosomes as inhalable carriers for enhancing its therapeutic efficacy via localized drug accumulation and addressing issues such as low bioavailability and severe toxicity. Methods: Niosomes were prepared by thin-film hydration method and were evaluated for in vitro therapeutic effectiveness in lung cancer cells. Results: The optimized niosomal formulation displayed optimized vesicle size, controlled and extended release of drug, and efficient aerodynamic properties indicating its suitability as an aerosolized formulation. In vitro studies revealed significantly superior cytotoxicity of nintedanib-loaded niosomes which was further validated by 3D spheroids. Conclusion: These findings establish the effectiveness of niosomes as inhalable delivery carriers which could serve as a promising strategy for delivery of nintedanib to treat several lung cancers.
Collapse
Affiliation(s)
- Snehal K Shukla
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St. John's University, NY 11439, USA
- Current Affiliation: Pfizer Worldwide R&D, Groton, CT 06340, USA
| | - Veronica Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St. John's University, NY 11439, USA
| | - Mimansa Goyal
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St. John's University, NY 11439, USA
| | - Vivek Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St. John's University, NY 11439, USA
| |
Collapse
|
19
|
Byun WS, Bae ES, Kim WK, Lee SK. Antitumor Activity of Rutaecarpine in Human Colorectal Cancer Cells by Suppression of Wnt/β-Catenin Signaling. JOURNAL OF NATURAL PRODUCTS 2022; 85:1407-1418. [PMID: 35544614 DOI: 10.1021/acs.jnatprod.2c00224] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Alkaloids derived from natural products have been traditionally used to treat various diseases, including cancers. Rutaecarpine (1), a β-carboline-type alkaloid obtained from Evodia rutaecarpa, has been previously reported as an anti-inflammatory agent. Nonetheless, its anticancer activity and the underlying molecular mechanisms remain to be explored. In the procurement of Wnt/β-catenin inhibitors from natural alkaloids, 1 was found to exhibit activity against the Wnt/β-catenin-response reporter gene. Since the abnormal activation of Wnt/β-catenin signaling is highly involved in colon carcinogenesis, the antitumor activity and molecular mechanisms of 1 were investigated in colorectal cancer (CRC) cells. The antiproliferative activity of 1 was associated with the suppression of the Wnt/β-catenin-mediated signaling pathway and its target gene expression in human CRC cells. 1 also induced G0/G1 cell cycle arrest and apoptotic cell death, and the antimigration and anti-invasion potential of 1 was confirmed through epithelial-mesenchymal transition biomarker inhibition by the regulation of Wnt signaling. The antitumor activity of 1 was supported in an Ls174T-implanted xenograft mouse model via Wnt target gene regulation. Overall, these findings suggest that targeting the Wnt/β-catenin signaling pathway by 1 is a promising therapeutic option for the treatment of human CRC harboring β-catenin mutation.
Collapse
Affiliation(s)
- Woong Sub Byun
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Seo Bae
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Won Kyung Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
20
|
Wei X, Zhang C, He S, Huang J, Huang J, Liew SS, Zeng Z, Pu K. A Dual‐locked Activatable Phototheranostic Probe for Biomarkers Regulated Photodynamic and Photothermal Cancer Therapy. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202202966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Xin Wei
- Nanyang Technological University School of chemical and biomedical Engineering SINGAPORE
| | - Chi Zhang
- Nanyang Technological University School of Chemical and Biomedical Engineering SINGAPORE
| | - Shasha He
- Nanyang Technological University School of Chemical and Biomedical Engineering SINGAPORE
| | - Jiaguo Huang
- Sun Yat-Sen University School of Pharmaceutical Sciences SINGAPORE
| | - Jingsheng Huang
- Nanyang Technological University School of Chemical and Biomedical Engineering SINGAPORE
| | - Si Si Liew
- Nanyang Technological University School of Chemical and Biomedical Engineering SINGAPORE
| | - Ziling Zeng
- Nanyang Technological University School of Chemical and Biomedical Engineering SINGAPORE
| | - Kanyi Pu
- Nanyang Technological University School of Chemical and Biomedical Engieering 70 Nanyang Drive 637457 Singapore SINGAPORE
| |
Collapse
|
21
|
Vazquez K, Saraswathibhatla A, Notbohm J. Effect of substrate stiffness on friction in collective cell migration. Sci Rep 2022; 12:2474. [PMID: 35169196 PMCID: PMC8847350 DOI: 10.1038/s41598-022-06504-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/25/2022] [Indexed: 11/09/2022] Open
Abstract
In collective cell migration, the motion results from forces produced by each cell and transmitted to the neighboring cells and to the substrate. Because inertia is negligible and the migration occurs over long time scales, the cell layer exhibits viscous behavior, where force and motion are connected by an apparent friction that results from the breaking and forming of adhesive bonds at the cell–cell and cell–substrate interfaces. Most theoretical models for collective migration include an apparent friction to connect force and motion, with many models making predictions that depend on the ratio of cell–cell and cell–substrate friction. However, little is known about factors that affect friction, leaving predictions of many theoretical models untested. Here, we considered how substrate stiffness and the number of adhesions affected friction at the cell–substrate interface. The experimental data were interpreted through prior theoretical models, which led to the same conclusion, that increased substrate stiffness increased the number of cell–substrate adhesions and caused increased cell–substrate friction. In turn, the friction affected the collective migration by altering the curvature at the edge of the cell layer. By revealing underlying factors affecting friction and demonstrating how friction perturbs the collective migration, this work provides experimental evidence supporting prior theoretical models and motivates the study of other ways to alter the collective migration by changing friction.
Collapse
Affiliation(s)
- Kelly Vazquez
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA.,Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Jacob Notbohm
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA. .,Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
22
|
Te Boekhorst V, Jiang L, Mählen M, Meerlo M, Dunkel G, Durst FC, Yang Y, Levine H, Burgering BMT, Friedl P. Calpain-2 regulates hypoxia/HIF-induced plasticity toward amoeboid cancer cell migration and metastasis. Curr Biol 2022; 32:412-427.e8. [PMID: 34883047 PMCID: PMC10439789 DOI: 10.1016/j.cub.2021.11.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 07/05/2021] [Accepted: 11/16/2021] [Indexed: 12/29/2022]
Abstract
Hypoxia, through hypoxia inducible factor (HIF), drives cancer cell invasion and metastatic progression in various cancer types. In epithelial cancer, hypoxia induces the transition to amoeboid cancer cell dissemination, yet the molecular mechanisms, relevance for metastasis, and effective intervention to combat hypoxia-induced amoeboid reprogramming remain unclear. Here, we identify calpain-2 as a key regulator and anti-metastasis target of hypoxia-induced transition from collective to amoeboid dissemination of breast and head and neck (HN) carcinoma cells. Hypoxia-induced amoeboid dissemination occurred through low extracellular matrix (ECM)-adhesive, predominantly bleb-based amoeboid movement, which was maintained by a low-oxidative and -glycolytic energy metabolism ("eco-mode"). Hypoxia induced calpain-2-mediated amoeboid conversion by deactivating β1 integrins through enzymatic cleavage of the focal adhesion adaptor protein talin-1. Consequently, targeted downregulation or pharmacological inhibition of calpain-2 restored talin-1 integrity and β1 integrin engagement and reverted amoeboid to elongated phenotypes under hypoxia. Calpain-2 activity was required for hypoxia-induced amoeboid conversion in the orthotopic mouse dermis and upregulated in invasive HN tumor xenografts in vivo, and attenuation of calpain activity prevented hypoxia-induced metastasis to the lungs. This identifies the calpain-2/talin-1/β1 integrin axis as a druggable mechanosignaling program that conserves energy yet enables metastatic dissemination that can be reverted by interfering with calpain activity.
Collapse
Affiliation(s)
- Veronika Te Boekhorst
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Cell Biology, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Liying Jiang
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marius Mählen
- Department of Cell Biology, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Maaike Meerlo
- Department of Molecular Cancer Research, Center for Molecular Medicine, UMC Utrecht, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands
| | - Gina Dunkel
- Department of Cell Biology, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Franziska C Durst
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yanjun Yang
- Center for Theoretical Biological Physics, Department of Applied Physics, Rice University, Houston, TX 77005, USA; Department of Physics, Northeastern University, Boston, MA 02115, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics, Department of Applied Physics, Rice University, Houston, TX 77005, USA; Department of Physics, Northeastern University, Boston, MA 02115, USA
| | - Boudewijn M T Burgering
- Department of Molecular Cancer Research, Center for Molecular Medicine, UMC Utrecht, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands; Cancer Genomics Center, 3584 CG Utrecht, the Netherlands
| | - Peter Friedl
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Cell Biology, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands; Cancer Genomics Center, 3584 CG Utrecht, the Netherlands.
| |
Collapse
|
23
|
Ohta S, Tago K, Kuchimaru T, Funakoshi-Tago M, Horie H, Aoki-Ohmura C, Matsugi J, Yanagisawa K. The role of MerTK in promoting cell migration is enhanced by the oncogenic Ras/IL-33 signaling axis. FEBS J 2021; 289:1950-1967. [PMID: 34743410 DOI: 10.1111/febs.16271] [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: 04/29/2021] [Revised: 09/15/2021] [Accepted: 11/05/2021] [Indexed: 11/29/2022]
Abstract
Ras genes are frequently mutated in many cancer types; however, there are currently no conclusively effective anticancer drugs against Ras-induced cancer. Therefore, the downstream effectors of Ras signaling need to be identified for the development of promising novel therapeutic approaches. We previously reported that oncogenic Ras induced the expression of NF-HEV/IL-33, a member of the interleukin-1 family, and showed that intracellular IL-33 was required for oncogenic Ras-induced cellular transformation. In the present study, we demonstrated that the c-Mer proto-oncogene tyrosine kinase (MerTK), a receptor tyrosine kinase, played essential roles in oncogenic Ras/IL-33 signaling. The expression of MerTK was enhanced in transformed NIH-3T3 cells by the expression of oncogenic Ras, H-Ras (G12V), in an IL-33-dependent manner. In human colorectal cancer tissues, MerTK expression also correlated with IL-33 expression. The knockdown of IL-33 or MerTK effectively attenuated the migration of NIH-3T3 cells transformed by H-Ras (G12V) and A549, LoVo, and HCT116 cells harboring an oncogenic K-Ras mutation. Furthermore, the suppression of Ras-induced cell migration by the knockdown of IL-33 was rescued by the enforced expression of MerTK. The present results also revealed that MerTK was effectively phosphorylated in NIH-3T3 cells transformed by Ras (G12V). Ras signaling was essential for the tyrosine phosphorylation of MerTK, and the kinase activity of MerTK was indispensable for accelerating cell migration. Collectively, the present results reveal a novel role for MerTK in cancer malignancy, which may be utilized to develop novel therapeutic strategies that target Ras-transformed cells.
Collapse
Affiliation(s)
- Satoshi Ohta
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan
| | - Kenji Tago
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan
| | | | | | - Hisanaga Horie
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | | | - Jitsuhiro Matsugi
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan
| | - Ken Yanagisawa
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
24
|
Wang L, Zhang D, Li J, Li F, Wei R, Jiang G, Xu H, Wang X, Zhou Y, Xi L. A novel ICG-labeled cyclic TMTP1 peptide dimer for sensitive tumor imaging and enhanced photothermal therapy in vivo. Eur J Med Chem 2021; 227:113935. [PMID: 34731764 DOI: 10.1016/j.ejmech.2021.113935] [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: 08/20/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/26/2022]
Abstract
TMTP1 is a polypeptide independently screened in our laboratory, which can target tumors in situ and metastases. In previous work, we have successfully developed a near-infrared (NIR) probe TMTP1-PEG4-ICG for tumor imaging. However, the limited ability to target tumor micrometastases hinders its further clinical application. Multimerization of peptides has been extensively demonstrated as an effective strategy to increase receptor binding affinity due to "multivalent effect" or "apparent cooperative affinity". In this study, a novel TMTP1 homodimer-directed NIR probe (TMTP1-PEG4)2-ICG was successfully constructed and synthesized. The cyclic TMTP1 peptides were bridged by two PEG4 linkers and then labeled with ICG-NHS for tumor imaging and photothermal therapy. In vivo biodistribution were assessed in normal BALB/c mice, and tumor targeting abilities of (TMTP1-PEG4)2-ICG and its monomer were evaluated and compared in 4T1-bearing subcutaneous tumor and lymph node metastasis model mice. Biodistribution analysis in vivo revealed that (TMTP1-PEG4)2-ICG was cleared mainly in both liver and kidney dependent way. Comparing with free ICG dye or TMTP1-PEG4-ICG probe, this improved (TMTP1-PEG4)2-ICG dimer showed more sensitive tumor imaging and could clearly identify tumors at a minimum volume of 10 mm3. Additionally, when compared to its monomer, lymph node (LN) metastases could also be apparently visualized and easily distinguished from normal LN by the novel dimer at 24 h post-injection. The blocking study revealed that the tumor accumulation of this probe was specifically medicated by receptor-ligand interaction. Furthermore, with the increase in stability and tumor targeting ability of ICG in vivo, the probe could also be an attractive photothermal agent to significantly inhibit tumor growth under 808 nm NIR laser irradiation. In conclusion, our work revealed that the novel (TMTP1-PEG4)2-ICG dimer could be a promising theranostic agent for sensitive tumor imaging and imaging-guided photothermal therapy, indicating its broad prospects for further clinical transformation.
Collapse
Affiliation(s)
- Ling Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Danya Zhang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jie Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Fei Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Rui Wei
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Guiying Jiang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Hanjie Xu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xueqian Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ying Zhou
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Ling Xi
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
25
|
Kozlovskaya V, Kharlampieva E. Anisotropic Particles through Multilayer Assembly. Macromol Biosci 2021; 22:e2100328. [PMID: 34644008 DOI: 10.1002/mabi.202100328] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/24/2021] [Indexed: 12/17/2022]
Abstract
The anisotropy in the shape of polymeric particles has been demonstrated to have many advantages over spherical particulates, including bio-mimetic behavior, shaped-directed flow, deformation, surface adhesion, targeting, motion, and permeability. The layer-by-layer (LbL) assembly is uniquely suited for synthesizing anisotropic particles as this method allows for simple and versatile replication of diverse colloid geometries with precise control over their chemical and physical properties. This review highlights recent progress in anisotropic particles of micrometer and nanometer sizes produced by a templated multilayer assembly of synthetic and biological macromolecules. Synthetic approaches to produce capsules and hydrogels utilizing anisotropic templates such as biological, polymeric, bulk hydrogel, inorganic colloids, and metal-organic framework crystals as sacrificial templates are overviewed. Structure-property relationships controlled by the anisotropy in particle shape and surface are discussed and compared with their spherical counterparts. Advances and challenges in controlling particle properties through varying shape anisotropy and surface asymmetry are outlined. The perspective applications of anisotropic colloids in biomedicine, including programmed behavior in the blood and tissues as artificial cells, nano-motors/sensors, and intelligent drug carriers are also discussed.
Collapse
Affiliation(s)
- Veronika Kozlovskaya
- Chemistry Department, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Eugenia Kharlampieva
- Chemistry Department, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.,UAB Center for Nanomaterials and Biointegration, UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
26
|
Liu Y, Zhao W, Cheng R, Hodgson J, Egan M, Pope CNC, Nikolinakos PG, Mao L. Simultaneous biochemical and functional phenotyping of single circulating tumor cells using ultrahigh throughput and recovery microfluidic devices. LAB ON A CHIP 2021; 21:3583-3597. [PMID: 34346469 DOI: 10.1039/d1lc00454a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Profiling circulating tumour cells (CTCs) in cancer patients' blood samples is critical to understand the complex and dynamic nature of metastasis. This task is challenged by the fact that CTCs are not only extremely rare in circulation but also highly heterogeneous in their molecular programs and cellular functions. Here we report a combinational approach for the simultaneous biochemical and functional phenotyping of patient-derived CTCs, using an integrated inertial ferrohydrodynamic cell separation (i2FCS) method and a single-cell microfluidic migration assay. This combinatorial approach offers unique capability to profile CTCs on the basis of their surface expression and migratory characteristics. We achieve this using the i2FCS method that successfully processes whole blood samples in a tumor cell marker and size agnostic manner. The i2FCS method enables an ultrahigh blood sample processing throughput of up to 2 × 105 cells s-1 with a blood sample flow rate of 60 mL h-1. Its short processing time (10 minutes for a 10 mL sample), together with a close-to-complete CTC recovery (99.70% recovery rate) and a low WBC contamination (4.07-log depletion rate by removing 99.992% of leukocytes), results in adequate and functional CTCs for subsequent studies in the single-cell migration device. For the first time, we employ this new approach to query CTCs with single-cell resolution in accordance with their expression of phenotypic surface markers and migration properties, revealing the dynamic phenotypes and the existence of a high-motility subpopulation of CTCs in blood samples from metastatic lung cancer patients. This method could be adopted to study the biological and clinical value of invasive CTC phenotypes.
Collapse
Affiliation(s)
- Yang Liu
- Department of Chemistry, The University of Georgia, Athens, Georgia, USA
| | - Wujun Zhao
- FCS Technology, LLC, Athens, GA, 30606, USA
| | - Rui Cheng
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, Georgia, USA.
| | - Jamie Hodgson
- University Cancer & Blood Center, LLC, Athens, GA, 30607, USA
| | - Mary Egan
- University Cancer & Blood Center, LLC, Athens, GA, 30607, USA
| | | | | | - Leidong Mao
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
27
|
Zhang M, Xu N, Xu W, Ling G, Zhang P. Potential therapies and diagnosis based on Golgi-targeted nano drug delivery systems. Pharmacol Res 2021; 175:105861. [PMID: 34464677 DOI: 10.1016/j.phrs.2021.105861] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
With the rapid development of nanotechnology, organelle-targeted nano drug delivery systems (NDDSs) have emerged as a potential method which can transport drugs specifically to the subcellular compartments like nucleus, mitochondrion, lysosome, endoplasmic reticulum (ER) and Golgi apparatus (GA). GA not only plays a key role in receiving, modifying, packaging and transporting proteins and lipids, but also contributes to a set of cellular processes. Golgi-targeted NDDSs can alter the morphology of GA and will become a promising strategy with high specificity, low-dose administration and decreased occurrence of side effects. In this review, Golgi-targeted NDDSs and their applications in disease therapies and diagnosis such as cancer, metastasis, fibrosis and neurological diseases are introduced. Meanwhile, modifications of NDDSs to achieve targeting strategies, Golgi-disturbing agents to change the morphology of GA, special endocytosis to achieve endosomal/lysosomal escape strategies are also involved.
Collapse
Affiliation(s)
- Manyue Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Na Xu
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Wenxin Xu
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Guixia Ling
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Peng Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
28
|
Chou SE, Lee KL, Wei PK, Cheng JY. Screening anti-metastasis drugs by cell adhesion-induced color change in a biochip. LAB ON A CHIP 2021; 21:2955-2970. [PMID: 34132296 DOI: 10.1039/d1lc00039j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Metastasis is a frequent complication of cancer and accounts for more than 60% of patients' mortality. Despite technological advancements, treatment options are still limited. Ion channels participate in the regulation of cell adhesion, whilst the regulation of cell adhesion further controls metastasis formation. However, to develop a new ion channel inhibitor targeting metastasis takes tremendous effort and resources; therefore, drug repurposing is an emerging strategy in oncology. In previous studies, we have developed a metal-based nanoslit surface plasmon resonance (SPR) platform to examine the influence of drugs on the cell adhesion process. In this work, we developed a scanner-based cell adhesion kinetic examination (CAKE) system that is capable of monitoring the cell adhesion process by measuring color changes of SPR biosensors. The system's performance was demonstrated by screening the anti-metastasis ability of compounds from a commercial ion-channel inhibitor library. Out of the 274 compounds from the inhibitor library, zinc pyrithione (ZPT) and terfenadine were demonstrated to influence CL1-5 cell adhesion. The cell responses to the two compounds were then compared with those by traditional cell adhesion assays where similar behavior was observed. Further investigation of the two compounds using wound healing and transwell assays was performed and inhibitions of both cell migration and invasion by the two compounds were also observed. The results indicate that ZPT and terfenadine are potential candidates for anti-metastasis drugs. Our work has demonstrated the label-free drug screening ability of our CAKE system for finding potential drugs for cancer treatment.
Collapse
Affiliation(s)
- Shih-En Chou
- Research Center for Applied Sciences, Academia Sinica Taiwan, Taipei, 11529, Taiwan.
| | - Kuang-Li Lee
- Research Center for Applied Sciences, Academia Sinica Taiwan, Taipei, 11529, Taiwan.
| | - Pei-Kuen Wei
- Research Center for Applied Sciences, Academia Sinica Taiwan, Taipei, 11529, Taiwan. and Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Ji-Yen Cheng
- Research Center for Applied Sciences, Academia Sinica Taiwan, Taipei, 11529, Taiwan. and Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan and Department of Mechanical and Mechatronic Engineering, National Taiwan Ocean University, Keelung, 20224, Taiwan and College of Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
| |
Collapse
|
29
|
The Influence of the Surface Topographical Cues of Biomaterials on Nerve Cells in Peripheral Nerve Regeneration: A Review. Stem Cells Int 2021; 2021:8124444. [PMID: 34349803 PMCID: PMC8328695 DOI: 10.1155/2021/8124444] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/05/2021] [Indexed: 01/01/2023] Open
Abstract
The surface topographies of artificial implants including surface roughness, surface groove size and orientation, and surface pore size and distribution have a great influence on the adhesion, migration, proliferation, and differentiation of nerve cells in the nerve regeneration process. Optimizing the surface topographies of biomaterials can be a key strategy for achieving excellent cell performance in various applications such as nerve tissue engineering. In this review, we offer a comprehensive summary of the surface topographies of nerve implants and their effects on nerve cell behavior. This review also emphasizes the latest work progress of the layered structure of the natural extracellular matrix that can be imitated by the material surface topology. Finally, the future development of surface topographies on nerve regeneration was prospectively remarked.
Collapse
|
30
|
Chen Z, Li Q, Xu S, Ouyang J, Wei H. Nanotopography-Modulated Epithelial Cell Collective Migration. J Biomed Nanotechnol 2021; 17:1079-1087. [PMID: 34167622 DOI: 10.1166/jbn.2021.3086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Matrix nanotopography plays an essential role in regulating cell behaviors including cell proliferation, differentiation, and migration. While studies on isolated single cell migration along the nanostructural orientation have been reported for various cell types, there remains a lack of understanding of how nanotopography regulates the behavior of collectively migrating cells during processes such as epithelial wound healing. We demonstrated that collective migration of epithelial cells was promoted on nanogratings perpendicular to, but not on those parallel to, the wound-healing axis. We further discovered that nanograting-modulated epithelial migration was dominated by the adhesion turnover process, which was Rho-associated protein kinase activity-dependent, and the lamellipodia protrusion at the cell leading edge, which was Rac1-GTPase activity-dependent. This work provides explanations to the distinct migration behavior of epithelial cells on nanogratings, and indicates that the effect of nanotopographic modulations on cell migration is cell-type dependent and involves complex mechanisms.
Collapse
Affiliation(s)
- Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Shihui Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Jun Ouyang
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Hongmei Wei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| |
Collapse
|
31
|
A predictive biomarker panel for bone metastases: Liquid biopsy approach. J Bone Oncol 2021; 29:100374. [PMID: 34189028 PMCID: PMC8220227 DOI: 10.1016/j.jbo.2021.100374] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 01/12/2023] Open
Abstract
Data mining of published microarray datasets directed us to the identification of a multi gene panel involving of 15 genes that are particular to bone metastases. Serum exosomal markers HSP90AA1, SPP1, IL3, and PTK2 found in the present study might be useful in detecting the early spread of bone metastases leading to better clinical outcomes. This multi-gene panel and their related pathways may assist as promising conclusion predictors using novel approaches of exosome as liquid biopsy and their application in therapeutic targets in breast and lung cancer patients with bone metastases.
Bone metastases is one of the common metastatic site and leading cause of cancer-related mortality in progressive cancer patients. The purpose of the present study is to establish a liquid biopsy based multi-gene classifier and associated signalling pathways for early diagnosis of bone metastases. We used publically available microarray datasets and analysed them in a platform/chip-specific manner using GeneSpring software. Analyses of gene expression datasets identified 15 consistently over-expressed genes with statistical significance. Further, expression profile of same set of 15 genes were compared in breast and lung cancer exosome derived mRNA with (n = 10) and without (n = 10) bone metastases against healthy controls. ROC curve analysis performed individually for all the 15 genes shortlisted the 5 most relevant genes with significant sensitivity and specificity in both cancers. This liquid biopsy-based bone metastases predictor using multi-gene panel is a unique approach with potential clinical applications for effective management of aggressive cancers.
Collapse
|
32
|
ECM Remodeling in Squamous Cell Carcinoma of the Aerodigestive Tract: Pathways for Cancer Dissemination and Emerging Biomarkers. Cancers (Basel) 2021. [DOI: 10.3390/cancers13112759
expr 955442319 + 839973387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Squamous cell carcinomas (SCC) include a number of different types of tumors developing in the skin, in hollow organs, as well as the upper aerodigestive tract (UADT) including the head and neck region and the esophagus which will be dealt with in this review. These tumors are often refractory to current therapeutic approaches with poor patient outcome. The most important prognostic determinant of SCC tumors is the presence of distant metastasis, significantly correlating with low patient survival rates. Rapidly emerging evidence indicate that the extracellular matrix (ECM) composition and remodeling profoundly affect SSC metastatic dissemination. In this review, we will summarize the current knowledge on the role of ECM and its remodeling enzymes in affecting the growth and dissemination of UADT SCC. Taken together, these published evidence suggest that a thorough analysis of the ECM composition in the UADT SCC microenvironment may help disclosing the mechanism of resistance to the treatments and help defining possible targets for clinical intervention.
Collapse
|
33
|
ECM Remodeling in Squamous Cell Carcinoma of the Aerodigestive Tract: Pathways for Cancer Dissemination and Emerging Biomarkers. Cancers (Basel) 2021; 13:cancers13112759. [PMID: 34199373 PMCID: PMC8199582 DOI: 10.3390/cancers13112759] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Local and distant metastasis of patients affected by squamous cell carcinoma of the upper aerodigestive tract predicts poor prognosis. In the latest years, the introduction of new therapeutic approaches, including targeted and immune therapies, has improved the overall survival. However, a large number of these patients do not benefit from these treatments. Thus, the identification of suitable prognostic and predictive biomarkers, as well as the discovery of new therapeutic targets have emerged as a crucial clinical need. In this context, the extracellular matrix represents a suitable target for the development of such therapeutic tools. In fact, the extracellular matrix is composed by complex molecules able to interact with a plethora of receptors and growth factors, thus modulating the dynamic crosstalk between cancer cells and the tumor microenvironment. In this review, we summarize the current knowledge of the role of the extracellular matrix in affecting squamous cell carcinoma growth and dissemination. Despite extracellular matrix is known to affect the development of many cancer types, only a restricted number of these molecules have been recognized to impact on squamous cell carcinoma progression. Thus, we consider that a thorough analysis of these molecules may be key to develop new potential therapeutic targets/biomarkers. Abstract Squamous cell carcinomas (SCC) include a number of different types of tumors developing in the skin, in hollow organs, as well as the upper aerodigestive tract (UADT) including the head and neck region and the esophagus which will be dealt with in this review. These tumors are often refractory to current therapeutic approaches with poor patient outcome. The most important prognostic determinant of SCC tumors is the presence of distant metastasis, significantly correlating with low patient survival rates. Rapidly emerging evidence indicate that the extracellular matrix (ECM) composition and remodeling profoundly affect SSC metastatic dissemination. In this review, we will summarize the current knowledge on the role of ECM and its remodeling enzymes in affecting the growth and dissemination of UADT SCC. Taken together, these published evidence suggest that a thorough analysis of the ECM composition in the UADT SCC microenvironment may help disclosing the mechanism of resistance to the treatments and help defining possible targets for clinical intervention.
Collapse
|
34
|
Gao J, Li S, Xu Q, Zhang X, Huang M, Dai X, Liu L. Exosomes Promote Pre-Metastatic Niche Formation in Gastric Cancer. Front Oncol 2021; 11:652378. [PMID: 34109113 PMCID: PMC8180914 DOI: 10.3389/fonc.2021.652378] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer has a high rate of metastasis, during which pre-metastatic niches (PMN) provide a supportive environment for the upcoming tumor cells. Exosomes are bilayer vesicles secreted by cells containing biological information that mediates communication between cells. Using exosomes, gastric cancer cells establish PMN remotely in multifarious perspectives, including immunosuppression, stroma remodeling, angiogenesis, mesothelial mesenchymal transformation, and organotropism. In turn, the cell components in PMN secrete exosomes that interact with each other and provide onco-promoting signals. In this review, we highlight the role of exosomes in PMN formation in gastric cancer and discuss their potential values in gastric cancer metastasis diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Jing Gao
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Song Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qian Xu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xue Zhang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Miao Huang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin Dai
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lian Liu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
35
|
Lin Q, Rong L, Jia X, Li R, Yu B, Hu J, Luo X, Badea SR, Xu C, Fu G, Lai K, Lee MC, Zhang B, Gong H, Zhou N, Chen XL, Lin SH, Fu G, Huang JD. IFN-γ-dependent NK cell activation is essential to metastasis suppression by engineered Salmonella. Nat Commun 2021; 12:2537. [PMID: 33953170 PMCID: PMC8099885 DOI: 10.1038/s41467-021-22755-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Metastasis accounts for 90% of cancer-related deaths and, currently, there are no effective clinical therapies to block the metastatic cascade. A need to develop novel therapies specifically targeting fundamental metastasis processes remains urgent. Here, we demonstrate that Salmonella YB1, an engineered oxygen-sensitive strain, potently inhibits metastasis of a broad range of cancers. This process requires both IFN-γ and NK cells, as the absence of IFN-γ greatly reduces, whilst depletion of NK cells in vivo completely abolishes, the anti-metastatic ability of Salmonella. Mechanistically, we find that IFN-γ is mainly produced by NK cells during early Salmonella infection, and in turn, IFN-γ promotes the accumulation, activation, and cytotoxicity of NK cells, which kill the metastatic cancer cells thus achieving an anti-metastatic effect. Our findings highlight the significance of a self-regulatory feedback loop of NK cells in inhibiting metastasis, pointing a possible approach to develop anti-metastatic therapies by harnessing the power of NK cells.
Collapse
Affiliation(s)
- Qiubin Lin
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,HKU-Zhejiang Institute of Research and Innovation (HKU-ZIRI), Hangzhou, China
| | - Li Rong
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Xian Jia
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China
| | - Renhao Li
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Bin Yu
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Jingchu Hu
- grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiao Luo
- grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - S. R. Badea
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Chen Xu
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Guofeng Fu
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China
| | - Kejiong Lai
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China
| | - Ming-chun Lee
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Baozhong Zhang
- grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huarui Gong
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Nan Zhou
- grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiao Lei Chen
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China ,grid.12955.3a0000 0001 2264 7233Cancer Research Center of Xiamen University, Xiamen, China
| | - Shu-hai Lin
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China
| | - Guo Fu
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China ,grid.12955.3a0000 0001 2264 7233Cancer Research Center of Xiamen University, Xiamen, China
| | - Jian-Dong Huang
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,HKU-Zhejiang Institute of Research and Innovation (HKU-ZIRI), Hangzhou, China ,grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
36
|
Liu Y, Zhao W, Cheng R, Harris BN, Murrow JR, Hodgson J, Egan M, Bankey A, Nikolinakos PG, Laver T, Meichner K, Mao L. Fundamentals of integrated ferrohydrodynamic cell separation in circulating tumor cell isolation. LAB ON A CHIP 2021; 21:1706-1723. [PMID: 33720269 PMCID: PMC8102387 DOI: 10.1039/d1lc00119a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Methods to separate circulating tumor cells (CTCs) from blood samples were intensively researched in order to understand the metastatic process and develop corresponding clinical assays. However current methods faced challenges that stemmed from CTCs' heterogeneity in their biological markers and physical morphologies. To this end, we developed integrated ferrohydrodynamic cell separation (iFCS), a scheme that separated CTCs independent of their surface antigen expression and physical characteristics. iFCS integrated both diamagnetophoresis of CTCs and magnetophoresis of blood cells together via a magnetic liquid medium, ferrofluid, whose magnetization could be tuned by adjusting its magnetic volume concentration. In this paper, we presented the fundamental theory of iFCS and its specific application in CTC separation. Governing equations of iFCS were developed to guide its optimization process. Three critical parameters that affected iFCS's cell separation performance were determined and validated theoretically and experimentally. These parameters included the sample flow rate, the volumetric concentration of magnetic materials in the ferrofluid, and the gradient of the magnetic flux density. We determined these optimized parameters in an iFCS device that led to a high recovery CTC separation in both spiked and clinical samples.
Collapse
Affiliation(s)
- Yang Liu
- Department of Chemistry, The University of Georgia, Athens, GA 30602, USA
| | - Wujun Zhao
- Department of Chemistry, The University of Georgia, Athens, GA 30602, USA
| | - Rui Cheng
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, GA 30602, USA.
| | - Bryana N Harris
- Department of Chemical Engineering, Auburn University, Auburn, AL 36830, USA
| | - Jonathan R Murrow
- Department of Medicine, Augusta University - The University of Georgia Medical Partnership, Athens, GA 30602, USA
| | - Jamie Hodgson
- University Cancer & Blood Center, LLC, Athens, GA 30607, USA
| | - Mary Egan
- University Cancer & Blood Center, LLC, Athens, GA 30607, USA
| | | | | | - Travis Laver
- Small Animal Medicine and Surgery, Veterinary Teaching Hospital, The University of Georgia, Athens, GA 30602, USA
| | - Kristina Meichner
- Department of Pathology, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA
| | - Leidong Mao
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
37
|
Role of AMPK and Akt in triple negative breast cancer lung colonization. Neoplasia 2021; 23:429-438. [PMID: 33839456 PMCID: PMC8042649 DOI: 10.1016/j.neo.2021.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/19/2022] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive disease with a 5-y relative survival rate of 11% after distant metastasis. To survive the metastatic cascade, tumor cells remodel their signaling pathways by regulating energy production and upregulating survival pathways. AMP-activated protein kinase (AMPK) and Akt regulate energy homeostasis and survival, however, the individual or synergistic role of AMPK and Akt isoforms during lung colonization by TNBC cells is unknown. The purpose of this study was to establish whether targeting Akt, AMPKα or both Akt and AMPKα isoforms in circulating cancer cells can suppress TNBC lung colonization. Transient silencing of Akt1 or Akt2 dramatically decreased metastatic colonization of lungs by inducing apoptosis or inhibiting invasion, respectively. Importantly, transient pharmacologic inhibition of Akt activity with MK-2206 or AZD5363 inhibitors did not prevent colonization of lung tissue by TNBC cells. Knockdown of AMPKα1, AMPKα2, or AMPKα1/2 also had no effect on metastatic colonization of lungs. Taken together, these findings demonstrate that transient decrease in AMPK isoforms expression alone or in combination with Akt1 in circulating tumor cells does not synergistically reduce TNBC metastatic lung colonization. Our results also provide evidence that Akt1 and Akt2 expression serve as a bottleneck that can challenge colonization of lungs by TNBC cells.
Collapse
|
38
|
A Cellular Potts Model for Analyzing Cell Migration across Constraining Pillar Arrays. AXIOMS 2021. [DOI: 10.3390/axioms10010032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Cell migration in highly constrained environments is fundamental in a wide variety of physiological and pathological phenomena. In particular, it has been experimentally shown that the migratory capacity of most cell lines depends on their ability to transmigrate through narrow constrictions, which in turn relies on their deformation capacity. In this respect, the nucleus, which occupies a large fraction of the cell volume and is substantially stiffer than the surrounding cytoplasm, imposes a major obstacle. This aspect has also been investigated with the use of microfluidic devices formed by dozens of arrays of aligned polymeric pillars that limit the available space for cell movement. Such experimental systems, in particular, in the designs developed by the groups of Denais and of Davidson, were here reproduced with a tailored version of the Cellular Potts model, a grid-based stochastic approach where cell dynamics are established by a Metropolis algorithm for energy minimization. The proposed model allowed quantitatively analyzing selected cell migratory determinants (e.g., the cell and nuclear speed and deformation, and forces acting at the nuclear membrane) in the case of different experimental setups. Most of the numerical results show a remarkable agreement with the corresponding empirical data.
Collapse
|
39
|
Lin F, Zhang C, Li Y, Yan J, Xu Y, Pan Y, Hu R, Liu L, Qu J. Human serum albumin gradient in serous ovarian cancer cryosections measured by fluorescence lifetime. BIOMEDICAL OPTICS EXPRESS 2021; 12:1195-1204. [PMID: 33796346 PMCID: PMC7984791 DOI: 10.1364/boe.415456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 05/05/2023]
Abstract
Human serum albumin (HSA) is a depot and carrier for many endogenous and exogenous molecules in blood. Many studies have demonstrated that the transport of HSA in tumor microenvironments contributes to tumor development and progression. In this report, we set up a multimodal nonlinear optical microscope system, combining two-photon excitation fluorescence, second harmonic generation, and two-photon fluorescence lifetime imaging microscopy. The fluorescence lifetime of a small squaraine dye (SD) is used to evaluate HSA concentrations in tumor tissue based on specific binding between SD and HSA. We used SD to stain the cryosections from serous ovarian cancer patients in high-grade (HGSOC) and low-grade (LGSOC), respectively, and found a gradient descent of HSA concentration from normal connective tissue to extracellular matrix to tumor masses from 13 to 2 µM for LGSOC patients and from 36 to 12 µM for HGSOC patients. We demonstrated that multimodal nonlinear optical microscopy can obtain similar results as those from traditional histologic staining, thus it is expected to move to clinical applications.
Collapse
Affiliation(s)
- Fangrui Lin
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province & Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Chenshuang Zhang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province & Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Yanping Li
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province & Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Junshuai Yan
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province & Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Yunjian Xu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province & Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Ying Pan
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Rui Hu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province & Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province & Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province & Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| |
Collapse
|
40
|
Cheng X, Cheng K. Visualizing cancer extravasation: from mechanistic studies to drug development. Cancer Metastasis Rev 2021; 40:71-88. [PMID: 33156478 PMCID: PMC7897269 DOI: 10.1007/s10555-020-09942-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
Metastasis is a multistep process that accounts for the majority of cancer-related death. By the end of metastasize dissemination, circulating tumor cells (CTC) need to extravasate the blood vessels at metastatic sites to form new colonization. Although cancer cell extravasation is a crucial step in cancer metastasis, it has not been successfully targeted by current anti-metastasis strategies due to the lack of a thorough understanding of the molecular mechanisms that regulate this process. This review focuses on recent progress in cancer extravasation visualization techniques, including the development of both in vitro and in vivo cancer extravasation models, that shed light on the underlying mechanisms. Specifically, multiple cancer extravasation stages, such as the adhesion to the endothelium and transendothelial migration, are successfully probed using these technologies. Moreover, the roles of different cell adhesive molecules, chemokines, and growth factors, as well as the mechanical factors in these stages are well illustrated. Deeper understandings of cancer extravasation mechanisms offer us new opportunities to escalate the discovery of anti-extravasation drugs and therapies and improve the prognosis of cancer patients.
Collapse
Affiliation(s)
- Xiao Cheng
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC, USA.
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA.
| |
Collapse
|
41
|
Hope JM, Bersi MR, Dombroski JA, Clinch AB, Pereles RS, Merryman WD, King MR. Circulating prostate cancer cells have differential resistance to fluid shear stress-induced cell death. J Cell Sci 2021; 134:jcs.251470. [PMID: 33526716 PMCID: PMC7929932 DOI: 10.1242/jcs.251470] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cells (CTCs) are exposed to fluid shear stress (FSS) of greater than 1000 dyn/cm2 (100 Pa) in circulation. Normally, CTCs that are exposed to FSS of this magnitude die. However, some CTCs develop resistance to this FSS, allowing them to colonize distant organs. We explored how prostate CTCs can resist cell death in response to forces of this magnitude. The DU145, PC3 and LNCaP human prostate cancer cell lines were used to represent cells of different metastatic origins. The cell lines were briefly treated with an average FSS of 3950 dyn/cm2 (395 Pa) using a 30 G needle and a syringe pump. DU145 cells had no change in cell viability, PC3 cells had some cell death and LNCaP cells exhibited significant cell death. These cell death responses correlated with increased cell membrane damage, less efficient membrane repair and increased stiffness. Additionally, FSS treatment prevented the LNCaP FSS-sensitive cell line from forming a growing tumor in vivo. This suggests that these properties play a role in FSS resistance and could represent potential targets for disrupting blood-borne metastasis. Summary: Prostate cancer cells have different sensitivities to fluid forces that alter their resistance to elevated blood flow-level fluid shear stress.
Collapse
Affiliation(s)
- Jacob M Hope
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Matthew R Bersi
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Jenna A Dombroski
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Andrea B Clinch
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Rebecca S Pereles
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| |
Collapse
|
42
|
Liang Z, Li X. Identification of ANXA1 as a potential prognostic biomarker and correlating with immune infiltrates in colorectal cancer. Autoimmunity 2021; 54:76-87. [PMID: 33596760 DOI: 10.1080/08916934.2021.1887148] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND ANXA1 is a calcium-dependent phospholipid-binding protein and is frequently associated with inflammation, cell proliferation and apoptosis. However, the relationship between ANXA1 and the prognosis of multiple tumours and tumour infiltrating immune cells remains unclear. METHODS Multivariate Cox proportional regression analysis was used for signature genes exploration in the basic of colon adenocarcinoma (COAD) RNA-sequence dataset obtained from TCGA, following the identification of 267 common differentially expressed genes, including ANXA1, among three expression profile datasets (GSE41328, GSE110224, and GSE113513). The differential expression of ANXA1 in different tumours and their corresponding normal tissues were evaluated through the Tumour Immune Estimation Resource (TIMER) and Oncomine database. Subsequently, we investigated the correlation between the expression level of ANXA1 and diverse panel of infiltrating immune cells and their related gene markers in colorectal cancer using correlation analysis in TIMER and GEPIA database. RESULTS The high expression of ANXA1 was demonstrated to be closely correlated with poor survival in patients with colorectal cancer. More importantly, we found that changes in ANXA1 expression showed a moderate to strong, and statistically significant, correlation with infiltrating levels of B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells. By contrast, there are only weak correlations between ANXA1 expression and immune cell infiltration in ESCA and STAD. ANXA1 expression was considerably associated with various immune markers involving immune cell recruitment, polarization of tumour-associated macrophages, and T cell exhaustion. CONCLUSION ANXA1 is not only an independent risk factor in the prediction of the prognosis of colorectal cancer, but also a crucial regulator in immune cell infiltration. This study may shed light on the clinical value of ANXA1, especially in the areas of early diagnosis of colorectal cancer and therapeutic target discovery.
Collapse
Affiliation(s)
- Zhikun Liang
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyan Li
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
43
|
Anti-Metastatic and Anti-Inflammatory Effects of Matrix Metalloproteinase Inhibition by Ginsenosides. Biomedicines 2021; 9:biomedicines9020198. [PMID: 33671187 PMCID: PMC7921986 DOI: 10.3390/biomedicines9020198] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/12/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are proteolytic enzymes which cleave extracellular matrix (ECM) and other substrates. They are deeply involved in both cancer metastasis and human chronic inflammatory diseases such as osteoarthritis and Crohn’s disease. Regulation of MMPs is closely associated with signaling molecules, especially mitogen-activated protein kinases (MAPKs), including three representative kinases, extracellular signal regulated kinases (ERK), p38 and c-Jun N-terminal kinases (JNK). Ginseng (Panax sp.) is a plant which has been traditionally used for medicinal applications. Ginsenosides are major metabolites which have potentials to treat various human diseases. In this review, the pharmacological effects of ginsenosides have been rigorously investigated; these include anti-metastatic and anti-inflammatory activities of ginsenosides associated with suppression of MMPs via regulation of various signaling pathways. This will highlight the importance of MMPs as therapeutic targets for anti-metastatic and anti-inflammatory drug development based on ginsenosides.
Collapse
|
44
|
Yang H, Kuo YH, Smith ZI, Spangler J. Targeting cancer metastasis with antibody therapeutics. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1698. [PMID: 33463090 DOI: 10.1002/wnan.1698] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022]
Abstract
Cancer metastasis, the spread of disease from a primary to a distal site through the circulatory or lymphatic systems, accounts for over 90% of all cancer related deaths. Despite significant progress in the field of cancer therapy in recent years, mortality rates remain dramatically higher for patients with metastatic disease versus those with local or regional disease. Although there is clearly an urgent need to develop drugs that inhibit cancer spread, the overwhelming majority of anticancer therapies that have been developed to date are designed to inhibit tumor growth but fail to address the key stages of the metastatic process: invasion, intravasation, circulation, extravasation, and colonization. There is growing interest in engineering targeted therapeutics, such as antibody drugs, that inhibit various steps in the metastatic cascade. We present an overview of antibody therapeutic approaches, both in the pipeline and in the clinic, that disrupt the essential mechanisms that underlie cancer metastasis. These therapies include classes of antibodies that indirectly target metastasis, including anti-integrin, anticadherin, and immune checkpoint blocking antibodies, as well as monoclonal and bispecific antibodies that are specifically designed to interrupt disease dissemination. Although few antimetastatic antibodies have achieved clinical success to date, there are many promising candidates in various stages of development, and novel targets and approaches are constantly emerging. Collectively, these efforts will enrich our understanding of the molecular drivers of metastasis, and the new strategies that arise promise to have a profound impact on the future of cancer therapeutic development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Huilin Yang
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yun-Huai Kuo
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zion I Smith
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jamie Spangler
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
45
|
Targetable Intercellular Signaling Pathways Facilitate Lung Colonization in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32767237 DOI: 10.1007/978-3-030-43085-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Outcomes for young people diagnosed with osteosarcoma hinge almost exclusively on whether they develop lung metastasis. The striking predilection that osteosarcoma shows for metastatic spread to lung suggests properties and/or lung interactions that generate tissue-specific survival and proliferation advantages. While these mechanisms remain overall poorly defined, studies have begun to describe biological elements important to metastasis. Mechanisms described to date include both cell-autonomous adaptations that allow disseminated tumor cells to survive the stressors imposed by metastasis and intercellular signaling networks that tumor cells exploit to pirate needed signals from surrounding tissues or to recruit other cells that create a more favorable niche. Evidence suggests that cell-autonomous changes are largely driven by epigenetic reprogramming of disseminated tumor cells that facilitates resistance to late apoptosis, manages endoplasmic reticulum (ER) stressors, promotes translation of complex transcripts, and activates clotting pathways. Tumor-host signaling pathways important for lung colonization drive interactions with lung epithelium, mesenchymal stem cells, and mediators of innate and adaptive immunity. In this chapter, we highlight one particular pathway that integrates cell-autonomous adaptations with lung-specific tumor-host interactions. In this mechanism, aberrant ΔNp63 expression primes tumor cells to produce IL6 and CXCL8 upon interaction with lung epithelial cells. This tumor-derived IL6 and CXCL8 then initiates autocrine, osteosarcoma-lung paracrine, and osteosarcoma-immune paracrine interactions that facilitate metastasis. Importantly, many of these pathways appear targetable with clinically feasible therapeutics. Ongoing work to better understand metastasis is driving efforts to improve outcomes by targeting the most devastating complication of this disease.
Collapse
|
46
|
Abstract
Transient receptor potential (TRP) channels comprise a diverse family of ion channels, the majority of which are calcium permeable and show sophisticated regulatory patterns in response to various environmental cues. Early studies led to the recognition of TRP channels as environmental and chemical sensors. Later studies revealed that TRP channels mediated the regulation of intracellular calcium. Mutations in TRP channel genes result in abnormal regulation of TRP channel function or expression, and interfere with normal spatial and temporal patterns of intracellular local Ca2+ distribution. The resulting dysregulation of multiple downstream effectors, depending on Ca2+ homeostasis, is associated with hallmarks of cancer pathophysiology, including enhanced proliferation, survival and invasion of cancer cells. These findings indicate that TRP channels affect multiple events that control cellular fate and play a key role in cancer progression. This review discusses the accumulating evidence supporting the role of TRP channels in tumorigenesis, with emphasis on prostate cancer. [BMB Reports 2020; 53(3): 125-132].
Collapse
Affiliation(s)
- Dongki Yang
- Departments of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Jaehong Kim
- Departments of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea
| |
Collapse
|
47
|
Wang X, Chen F, Gou S. Anti-tumor effects and cell motility inhibition of the DN604-gemcitabine combined treatment in human bladder cancer models. Bioorg Med Chem 2020; 29:115858. [PMID: 33218897 DOI: 10.1016/j.bmc.2020.115858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/20/2020] [Accepted: 11/01/2020] [Indexed: 01/20/2023]
Abstract
Bladder cancer is one of the major tumors for men in the world, in which therapy the combination of cisplatin and gemcitabine is still fist-line applied to treat with advanced or metastatic bladder cancer. In our early study, we developed a potential Pt(II) agent, DN604, which has anti-tumor effect as potent as cisplatin toward bladder cancers. Herein, we aim at investigating the combinatory application of DN604 with gemcitabine for bladder cancer treatment. In vitro studies proved that the combined treatment of DN604 and gemcitabine could limit cell proliferation by elevating the incidence of DNA damage induced apoptosis. Notably, further researches showed that the DN604-gemcitabine treatment suppressed cell autophagy to inhibit cell motility upon the ROS dependent p38 MAPK signaling pathway, explicating its better anti-tumor activity than single drug treatment or the cisplatin-gemcitabine treatment. In vivo tests confirmed that the DN604-gemcitabine treatment has superior anti-tumor activity with low toxicity to cisplatin or its combination with gemcitabine treatments. DN604 plus gemcitabine, is of great significance for the treatment with human bladder cancer. Our study has provided a potential combination treatment option.
Collapse
Affiliation(s)
- Xinyi Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Feihong Chen
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China.
| |
Collapse
|
48
|
Doi T, Ogawa H, Tanaka Y, Hayashi Y, Maniwa Y. Bex1 significantly contributes to the proliferation and invasiveness of malignant tumor cells. Oncol Lett 2020; 20:362. [PMID: 33133262 PMCID: PMC7590424 DOI: 10.3892/ol.2020.12226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/15/2020] [Indexed: 01/08/2023] Open
Abstract
Invasion has a significant role in cancer progression, including expansion to surrounding tissue and metastasis. Previously, we assessed the invasive ability of cancer cells using an easy-to-prepare double-layered collagen gel hemisphere (DL-CGH) method by which cancer cell invasion can be easily visualized. The present study examined multiple lung adenocarcinoma and malignant pleural mesothelioma (MPM) cell lines using the DL-CGH method and identified inherently invasive cell lines. Next, by comparing gene expression between invasive and non-invasive cells by cDNA microarray, the potential candidate gene brain-expressed x-linked protein 1 (Bex1) was identified to be involved in cancer invasion, as it was highly expressed in the invasive cell lines. Downregulation of Bex1 suppressed the invasion and proliferation of the invasive tumor cell lines. The findings of the present study suggested that Bex1 may promote metastasis in vivo and could be a potential oncogene and molecular therapeutic target in lung adenocarcinoma and MPM.
Collapse
Affiliation(s)
- Takefumi Doi
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Hiroyuki Ogawa
- Department of Thoracic Surgery, Hyogo Cancer Center, Akashi, Hyogo 673-8558, Japan
| | - Yugo Tanaka
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Yoshitake Hayashi
- Division of Molecular Medicine and Medical Genetics, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Yoshimasa Maniwa
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| |
Collapse
|
49
|
Breast Cancer-Derived Microparticles Reduce Cancer Cell Adhesion, an Effect Augmented by Chemotherapy. Cells 2020; 9:cells9102269. [PMID: 33050539 PMCID: PMC7650796 DOI: 10.3390/cells9102269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/08/2020] [Accepted: 10/10/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor cell heterogeneity is primarily dictated by mutational changes, sometimes leading to clones that undergo a metastatic switch. However, little is known about tumor heterogeneity following chemotherapy perturbation. Here we studied the possible involvement of tumor-derived extracellular vesicles, often referred to as tumor-derived microparticles (TMPs), as mediators of the metastatic switch in the tumor microenvironment by hindering cell adhesion properties. Specifically, we show that highly metastatic or chemotherapy-treated breast cancer cells shed an increased number of TMPs compared to their respective controls. We found that these TMPs substantially reduce cell adhesion and disrupt actin filament structure, therefore increasing their biomechanical force pace, further implicating tumor cell dissemination as part of the metastatic cascade. Our results demonstrate that these pro-metastatic effects are mediated in part by CD44 which is highly expressed in TMPs obtained from highly metastatic cells or cells exposed to chemotherapy when compared to cells with low metastatic potential. Consequently, when we inhibited CD44 expression on TMPs by a pharmacological or a genetic approach, increased tumor cell adhesion and re-organized actin filament structure were observed. We also demonstrated that breast cancer patients treated with paclitaxel chemotherapy exhibited increased CD44-expressing TMPs. Overall, our study provides further insights into the role of TMPs in promoting metastasis, an effect which is augmented when tumor cells are exposed to chemotherapy.
Collapse
|
50
|
Chen SR, Wang SW, Sheu JH, Chang TH, Cheng YB. Zoanthamine Alkaloid Derivatives from the Zoantharian Zoanthus vietnamensis with Antimetastatic Activity. J Org Chem 2020; 85:12553-12560. [DOI: 10.1021/acs.joc.0c01731] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Shu-Rong Chen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Shih-Wei Wang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Jyh-Horng Sheu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Ting-Hsuan Chang
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yuan-Bin Cheng
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| |
Collapse
|