1
|
Kofman K, Levin M. Bioelectric pharmacology of cancer: A systematic review of ion channel drugs affecting the cancer phenotype. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 191:25-39. [PMID: 38971325 DOI: 10.1016/j.pbiomolbio.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/21/2024] [Accepted: 07/04/2024] [Indexed: 07/08/2024]
Abstract
Cancer is a pernicious and pressing medical problem; moreover, it is a failure of multicellular morphogenesis that sheds much light on evolutionary developmental biology. Numerous classes of pharmacological agents have been considered as cancer therapeutics and evaluated as potential carcinogenic agents; however, these are spread throughout the primary literature. Here, we briefly review recent work on ion channel drugs as promising anti-cancer treatments and present a systematic review of the known cancer-relevant effects of 109 drugs targeting ion channels. The roles of ion channels in cancer are consistent with the importance of bioelectrical parameters in cell regulation and with the functions of bioelectric signaling in morphogenetic signals that act as cancer suppressors. We find that compounds that are well-known for having targets in the nervous system, such as voltage-gated ion channels, ligand-gated ion channels, proton pumps, and gap junctions are especially relevant to cancer. Our review suggests further opportunities for the repurposing of numerous promising candidates in the field of cancer electroceuticals.
Collapse
Affiliation(s)
- Karina Kofman
- Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Michael Levin
- Allen Discovery Center at Tufts University, USA; Wyss Institute for Biologically Inspired Engineering at Harvard University, USA.
| |
Collapse
|
2
|
Shao Z, Wang B, Gao H, Zhang S. Microenvironmental interference with intra-articular stem cell regeneration influences the onset and progression of arthritis. Front Genet 2024; 15:1380696. [PMID: 38841721 PMCID: PMC11150611 DOI: 10.3389/fgene.2024.1380696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Studies have indicated that the preservation of joint health and the facilitation of damage recovery are predominantly contingent upon the joint's microenvironment, including cell-cell interactions, the extracellular matrix's composition, and the existence of local growth factors. Mesenchymal stem cells (MSCs), which possess the capacity to self-renew and specialize in many directions, respond to cues from the microenvironment, and aid in the regeneration of bone and cartilage, are crucial to this process. Changes in the microenvironment (such as an increase in inflammatory mediators or the breakdown of the extracellular matrix) in the pathological context of arthritis might interfere with stem cell activation and reduce their ability to regenerate. This paper investigates the potential role of joint microenvironmental variables in promoting or inhibiting the development of arthritis by influencing stem cells' ability to regenerate. The present status of research on stem cell activity in the joint microenvironment is also outlined, and potential directions for developing new treatments for arthritis that make use of these intervention techniques to boost stem cell regenerative potential through altering the intra-articular environment are also investigated. This review's objectives are to investigate these processes, offer fresh perspectives, and offer a solid scientific foundation for the creation of arthritic treatment plans in the future.
Collapse
Affiliation(s)
| | | | | | - Shenqi Zhang
- Department of Joint and Sports Medicine, Zaozhuang Municipal Hospital Affiliated to Jining Medical University, Zaozhuang, Shandong, China
| |
Collapse
|
3
|
Claes J, Agten A, Blázquez-Moreno A, Crabbe M, Tuefferd M, Goehlmann H, Geys H, Peng CY, Neyens T, Faes C. The influence of resolution on the predictive power of spatial heterogeneity measures as biomarkers of liver fibrosis. Comput Biol Med 2024; 171:108231. [PMID: 38422965 DOI: 10.1016/j.compbiomed.2024.108231] [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: 10/09/2023] [Revised: 01/23/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Spatial heterogeneity of cells in liver biopsies can be used as biomarker for disease severity of patients. This heterogeneity can be quantified by non-parametric statistics of point pattern data, which make use of an aggregation of the point locations. The method and scale of aggregation are usually chosen ad hoc, despite values of the aforementioned statistics being heavily dependent on them. Moreover, in the context of measuring heterogeneity, increasing spatial resolution will not endlessly provide more accuracy. The question then becomes how changes in resolution influence heterogeneity indicators, and subsequently how they influence their predictive abilities. In this paper, cell level data of liver biopsy tissue taken from chronic Hepatitis B patients is used to analyze this issue. Firstly, Morisita-Horn indices, Shannon indices and Getis-Ord statistics were evaluated as heterogeneity indicators of different types of cells, using multiple resolutions. Secondly, the effect of resolution on the predictive performance of the indices in an ordinal regression model was investigated, as well as their importance in the model. A simulation study was subsequently performed to validate the aforementioned methods. In general, for specific heterogeneity indicators, a downward trend in predictive performance could be observed. While for local measures of heterogeneity a smaller grid-size is outperforming, global measures have a better performance with medium-sized grids. In addition, the use of both local and global measures of heterogeneity is recommended to improve the predictive performance.
Collapse
Affiliation(s)
- Jari Claes
- Data Science Institute, UHasselt - Hasselt University, Agoralaan 1, Diepenbeek, 3590, Belgium.
| | - Annelies Agten
- Data Science Institute, UHasselt - Hasselt University, Agoralaan 1, Diepenbeek, 3590, Belgium
| | - Alfonso Blázquez-Moreno
- Discovery Statistics, Global Development, Janssen Research and Development, Turnhoutseweg 30, Beerse, 2340, Belgium
| | - Marjolein Crabbe
- Discovery Statistics, Global Development, Janssen Research and Development, Turnhoutseweg 30, Beerse, 2340, Belgium
| | - Marianne Tuefferd
- Translational Biomarkers, Infectious Diseases, Janssen Research and Development, Turnhoutseweg 30, Beerse, 2340, Belgium
| | - Hinrich Goehlmann
- Translational Biomarkers, Infectious Diseases, Janssen Research and Development, Turnhoutseweg 30, Beerse, 2340, Belgium
| | - Helena Geys
- Discovery Statistics, Global Development, Janssen Research and Development, Turnhoutseweg 30, Beerse, 2340, Belgium
| | | | - Thomas Neyens
- Data Science Institute, UHasselt - Hasselt University, Agoralaan 1, Diepenbeek, 3590, Belgium; L-BioStat, KU Leuven, Kapucijnenvoer 35, Leuven, 3000, Belgium
| | - Christel Faes
- Data Science Institute, UHasselt - Hasselt University, Agoralaan 1, Diepenbeek, 3590, Belgium
| |
Collapse
|
4
|
Lou E, Clemente V, Grube M, Svedbom A, Nelson AC, Blome F, Staebler A, Kommoss S, Bazzaro M. Tumor-Stroma Proportion to Predict Chemoresistance in Patients With Ovarian Cancer. JAMA Netw Open 2024; 7:e240407. [PMID: 38411963 PMCID: PMC10900967 DOI: 10.1001/jamanetworkopen.2024.0407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/07/2024] [Indexed: 02/28/2024] Open
Abstract
IMPORTANCE Platinum-based chemotherapy is the backbone of standard-of-care treatment for patients with advanced-stage, high-grade serous carcinoma (HGSC), the most common form of ovarian cancer; however, one-third of patients have or acquire chemoresistance toward platinum-based therapies. OBJECTIVE To demonstrate the utility of tumor-stroma proportion (TSP) as a predictive biomarker of chemoresistance of HGSC, progression-free survival (PFS), and overall survival (OS). DESIGN, SETTING, AND PARTICIPANTS This prognostic study leveraged tumors from patients with HGSC in The Cancer Genome Atlas (TCGA) cohort (1993-2013) and an independent cohort of resected clinical specimens from patients with HGSC (2004-2014) available in diagnostic and tissue microarray formats from the University of Tübingen in Germany. Data analysis was conducted from January 2021 to January 2024. EXPOSURE Diagnosis of HGSC. MAIN OUTCOMES AND MEASURES Principal outcome measures were the ability of TSP to predict platinum chemoresistance, PFS, and OS. Using hematoxylin and eosin-stained slides from the Tübingen cohort (used for routine diagnostic assessment from surgical specimens) as well as tissue microarrays, representative sections of tumors for scoring of TSP were identified using previously evaluated cutoffs of 50% stroma or greater (high TSP) and less than 50% stroma (low TSP). Digitized slides from the TCGA Cohort were analyzed and scored in a similar fashion. Kaplan-Meier time-to-event functions were fit to estimate PFS and OS. RESULTS The study included 103 patients (mean [SD] age, 61.6 [11.1] years) from the TCGA cohort and 192 patients (mean [SD] age at diagnosis, 63.7 [11.1] years) from the Tübingen cohort. In the TCGA cohort, there was no significant association of TSP levels with chemoresistance, PFS, or OS. However, in the Tübingen cohort, high TSP was associated with significantly shorter PFS (HR, 1.586; 95% CI, 1.093-2.302; P = .02) and OS (hazard ratio [HR], 1.867; 1.249-2.789; P = .002). Patients with chemoresistant tumors were twice as likely to have high TSP as compared to patients with chemosensitive tumors (HR, 2.861; 95% CI, 1.256-6.515; P = .01). In tissue microarrays from 185 patients from the Tübingen cohort, high TSP was again associated with significantly shorter PFS (HR, 1.675; 95% CI, 1.012-2.772 P = .04) and OS (HR, 2.491; 95% CI, 1.585-3.912; P < .001). CONCLUSIONS AND RELEVANCE In this prognostic study, TSP was a consistent and reproducible marker of clinical outcome measures of HGSC, including PFS, OS, and platinum chemoresistance. Accurate and cost-effective predictive biomarkers of platinum chemotherapy resistance are needed to identify patients most likely to benefit from standard treatments, and TSP can easily be implemented and integrated into prospective clinical trial design and adapted to identify patients who are least likely to benefit long-term from conventional platinum-based cytotoxic chemotherapy treatment at the time of initial diagnosis.
Collapse
Affiliation(s)
- Emil Lou
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis
| | | | - Marcel Grube
- Department of Women’s Health, Tübingen University Hospital, Tübingen, Germany
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis
| | - Axel Svedbom
- Division of Dermatology and Venereology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Andrew C. Nelson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Freya Blome
- Institute of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Annette Staebler
- Institute of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Stefan Kommoss
- Department of Women’s Health, Tübingen University Hospital, Tübingen, Germany
| | | |
Collapse
|
5
|
Grasso G, Colella F, Forciniti S, Onesto V, Iuele H, Siciliano AC, Carnevali F, Chandra A, Gigli G, Del Mercato LL. Fluorescent nano- and microparticles for sensing cellular microenvironment: past, present and future applications. NANOSCALE ADVANCES 2023; 5:4311-4336. [PMID: 37638162 PMCID: PMC10448310 DOI: 10.1039/d3na00218g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/13/2023] [Indexed: 08/29/2023]
Abstract
The tumor microenvironment (TME) demonstrates distinct hallmarks, including acidosis, hypoxia, reactive oxygen species (ROS) generation, and altered ion fluxes, which are crucial targets for early cancer biomarker detection, tumor diagnosis, and therapeutic strategies. Various imaging and sensing techniques have been developed and employed in both research and clinical settings to visualize and monitor cellular and TME dynamics. Among these, ratiometric fluorescence-based sensors have emerged as powerful analytical tools, providing precise and sensitive insights into TME and enabling real-time detection and tracking of dynamic changes. In this comprehensive review, we discuss the latest advancements in ratiometric fluorescent probes designed for the optical mapping of pH, oxygen, ROS, ions, and biomarkers within the TME. We elucidate their structural designs and sensing mechanisms as well as their applications in in vitro and in vivo detection. Furthermore, we explore integrated sensing platforms that reveal the spatiotemporal behavior of complex tumor cultures, highlighting the potential of high-resolution imaging techniques combined with computational methods. This review aims to provide a solid foundation for understanding the current state of the art and the future potential of fluorescent nano- and microparticles in the field of cellular microenvironment sensing.
Collapse
Affiliation(s)
- Giuliana Grasso
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Francesco Colella
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
- Department of Mathematics and Physics ''Ennio De Giorgi", University of Salento c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Stefania Forciniti
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Helena Iuele
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Anna Chiara Siciliano
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
- Department of Mathematics and Physics ''Ennio De Giorgi", University of Salento c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Federica Carnevali
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
- Department of Mathematics and Physics ''Ennio De Giorgi", University of Salento c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Anil Chandra
- Centre for Research in Pure and Applied Sciences, Jain (Deemed-to-be-university) Bangalore Karnataka 560078 India
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
- Department of Mathematics and Physics ''Ennio De Giorgi", University of Salento c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| |
Collapse
|
6
|
Lou E, Clemente V, Grube M, Svedbom A, Nelson A, Blome F, Staebler A, Kommoss S, Bazzaro M. Clinical Significance of the Stromatic Component in Ovarian Cancer: Quantity Over Quality in Outcome Prediction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546712. [PMID: 37425832 PMCID: PMC10326982 DOI: 10.1101/2023.06.27.546712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Background The tumor stroma is composed of a complex network of non-cancerous cells and extracellular matrix elements that collectively are crucial for cancer progression and treatment response. Within the realm of ovarian cancer, the expression of the stromal gene cluster has been linked to poorer progression-free and overall survival rates. However, in the age of precision medicine and genome sequencing, the notion that the simple measurement of tumor-stroma proportion alone can serve as a biomarker for clinical outcome is a topic that continues to generate controversy and provoke discussion. Our current study reveals that it is the quantity of stroma, rather than its quality, that serves as a clinically significant indicator of patient outcome in ovarian cancer. Methods This study leveraged the High-Grade-Serous-Carcinoma (HGSC) cohort of the publicly accessible Cancer Genome Atlas Program (TCGA) along with an independent cohort comprising HGSC clinical specimens in diagnostic and Tissue Microarray formats. Our objective was to investigate the correlation between the Tumor-Stroma-Proportion (TSP) and progression-free survival (PFS), overall survival (OS), and response to chemotherapy. We assessed these associations using H&E-stained slides and tissue microarrays. Our analysis employed semi-parametric models that accounted for age, metastases, and residual disease as controlling factors. Results We found that high TSP (>50% stroma) was associated with significantly shorter progression-free survival (PFS) (p=0.016) and overall survival (OS) (p=0.006). Tumors from patients with chemoresistant tumors were twice as likely to have high TSP as compared to tumors from chemosensitive patients (p=0.012). In tissue microarrays, high TSP was again associated with significantly shorter PFS (p=0.044) and OS (p=0.0001), further confirming our findings. The Area Under the ROC curve for the model predicting platinum was estimated at 0.7644. Conclusions In HGSC, TSP was a consistent and reproducible marker of clinical outcome measures, including PFS, OS, and platinum chemoresistance. Assessment of TSP as a predictive biomarker that can be easily implemented and integrated into prospective clinical trial design and adapted to identify, at time of initial diagnosis, patients who are least likely to benefit long-term from conventional platinum-based cytotoxic chemotherapy treatment.
Collapse
|
7
|
Yang X, Xu G, Liu X, Zhou G, Zhang B, Wang F, Wang L, Li B, Li L. Carbon nanomaterial-involved EMT and CSC in cancer. REVIEWS ON ENVIRONMENTAL HEALTH 2023; 38:1-13. [PMID: 34619029 DOI: 10.1515/reveh-2021-0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/26/2021] [Indexed: 06/13/2023]
Abstract
Carbon nanomaterials (CNMs) are ubiquitous in our daily lives because of the outstanding physicochemical properties. CNMs play curial parts in industrial and medical fields, however, the risks of CNMs exposure to human health are still not fully understood. In view of, it is becoming extremely difficult to ignore the existence of the toxicity of CNMs. With the increasing exploitation of CNMs, it's necessary to evaluate the potential impact of these materials on human health. In recent years, more and more researches have shown that CNMs are contributed to the cancer formation and metastasis after long-term exposure through epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs) which is associated with cancer progression and invasion. This review discusses CNMs properties and applications in industrial and medical fields, adverse effects on human health, especially the induction of tumor initiation and metastasis through EMT and CSCs procedure.
Collapse
Affiliation(s)
- Xiaotong Yang
- Tianjin Medical University General Hospital, Tianjin, China
| | - Gongquan Xu
- Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaolong Liu
- Tianjin Medical University General Hospital, Tianjin, China
| | - Guiming Zhou
- Tianjin Medical University General Hospital, Tianjin, China
| | - Bing Zhang
- Rushan Hospital of Traditional Chinese Medicine, Weihai, China
| | - Fan Wang
- Tianjin Medical University General Hospital, Tianjin, China
| | - Lingjuan Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Bin Li
- Tianjin Medical University General Hospital, Tianjin, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Liming Li
- Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
8
|
Ruggiero A, Triarico S, Romano A, Maurizi P, Attina G, Mastrangelo S. Bisphosphonates: From Pharmacology to Treatment. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2023; 16:221-229. [DOI: 10.13005/bpj/2603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Bisphosphonates are among the most widely used drugs in the world for their many clinical indications. Their mechanism of action is based on the increase in the level of bone mineralization through the inhibition of osteoclastic activity and the induction of osteoblastic activity. Recent studies also attribute to bisphosphonates an antineoplastic activity, due to the ability of these drugs to inhibit neo angiogenesis, inhibiting the proliferation of endothelial cells. Bisphosphonates have several common properties, including poorly absorbed orally, high affinity for bone mineral, inhibitory effects on osteoclastic bone resorption, prolonged bone retention, and elimination in the urine. Bisphosphonates are generally well tolerated but their use can be, however, burdened by serious side effects such as hypocalcaemia, renal impairment, and aseptic osteonecrosis of the jaw.
Collapse
Affiliation(s)
- Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Alberto Romano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Giorgio Attina
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
9
|
Taha M, Rahim F, Khan IU, Uddin N, Farooq RK, Wadood A, Rehman AU, Khan KM. Synthesis of thiazole-based-thiourea analogs: as anticancer, antiglycation and antioxidant agents, structure activity relationship analysis and docking study. J Biomol Struct Dyn 2023; 41:12077-12092. [PMID: 36695088 DOI: 10.1080/07391102.2023.2171134] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/27/2022] [Indexed: 01/26/2023]
Abstract
This work reports the convenient approach for the synthesis of thiazole based thiourea derivatives (1-21) from 2-bromo-1-(4-fluorophenyl)thiazole-1-one and phenyl isothiocyanates. The scope and diversity were achieved from readily available phenyl isothiocyanates. This protocol involves an oxidative C-S bond formation. Moreover, hybrid thiazole based thiourea scaffolds (1-21) according to literature known protocol were screened in vitro for anticancer Potential against breast cancer, antiglycation and antioxidant inhibitory profile. All newly developed scaffolds were showed moderate to good inhibitory potentials ranging from 0.10 ± 0.01 µM to 11.40 ± 0.20 µM, 64.20 ± 0.40 µM to 385.10 ± 1.70 µM and 8.90 ± 0.20 µM to 39.20 ± 0.50 µM against anticancer, antiglycation and antioxidant respectively. Among the series, compounds 12 (IC50 = 0.10 ± 0.01 µM), 10 (IC50 = 64.20 ± 0.40 µM) and 12 (IC50 = 8.90 ± 0.20 µM) with flouro substitution at phenyl ring of thiourea were identified to be the most potent among the series having excellent anticancer, antiglycation and antioxidant potential. The structure of all the newly synthetics scaffolds were confirmed by using different types of spectroscopic techniques such as HREI-MS, 1H- and 13C-NMR spectroscopy. To find structure-activity relationship, molecular docking studies were carried out to understand the binding mode of active inhibitors with active site of enzymes and results supported the experimental data.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Muhammad Taha
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra, Khyber Pakhtunkhwa, Pakistan
| | - Ihsan Ullah Khan
- Department of Chemistry, Hazara University, Mansehra, Khyber Pakhtunkhwa, Pakistan
| | - Nizam Uddin
- Department of Chemistry, University of Karachi, Karachi, Pakistan
| | - Rai Khalid Farooq
- Department of Neuroscience Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Ashfaq Ur Rehman
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
10
|
Liu QP, Chen YY, An P, Rahman K, Luan X, Zhang H. Natural products targeting macrophages in tumor microenvironment are a source of potential antitumor agents. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154612. [PMID: 36610172 DOI: 10.1016/j.phymed.2022.154612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Macrophages are one of the major cell types in the immune system and are closely related to tumor development, which can be polarized into M1 type with anti-tumor activity or M2 type with pro-tumor activity. The infiltration of more macrophages into tumor predicts poorer prognosis due to their more exhibition of M2 phenotype under the influence of many factors in the tumor microenvironment (TME). Therefore, reverse of M2 macrophage polarization in TME is conducive to the suppression of tumor deterioration and understanding the influencing factors of macrophage polarization is helpful to provide new ideas for the subsequent targeting macrophages for tumor therapy. PURPOSE This review summarizes the effects of TME on macrophage polarization and natural products against M2 macrophage polarization, which may provide some directions for tumor therapy. METHODS The search of relevant literature was conducted using the PubMed, Science Direct, CNKI and Web of Science databases with the search terms "macrophage", "tumor microenvironment", "natural product" and "tumor". RESULTS The mutual transformation of M1 and M2 phenotypes in macrophages is influenced by many factors. Tumor cells affect the polarization of macrophages by regulating the expression of genes and proteins and the secretion of cytokines. The expression of some genes or proteins in macrophages is also related to their own polarization. Many natural products can reverse M2 polarization of macrophages which has been summarized in this review. CONCLUSION Regulation of macrophage polarization in TME can inhibit tumor development, and natural products have the potential to impede tumor development by regulating macrophage polarization.
Collapse
Affiliation(s)
- Qiu-Ping Liu
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu-Ying Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Pei An
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Khalid Rahman
- School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
11
|
Shi T, Yu H, Blair RH. Integrated regulatory and metabolic networks of the tumor microenvironment for therapeutic target prioritization. Stat Appl Genet Mol Biol 2023; 22:sagmb-2022-0054. [PMID: 37988745 DOI: 10.1515/sagmb-2022-0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 09/28/2023] [Indexed: 11/23/2023]
Abstract
Translation of genomic discovery, such as single-cell sequencing data, to clinical decisions remains a longstanding bottleneck in the field. Meanwhile, computational systems biological models, such as cellular metabolism models and cell signaling pathways, have emerged as powerful approaches to provide efficient predictions in metabolites and gene expression levels, respectively. However, there has been limited research on the integration between these two models. This work develops a methodology for integrating computational models of probabilistic gene regulatory networks with a constraint-based metabolism model. By using probabilistic reasoning with Bayesian Networks, we aim to predict cell-specific changes under different interventions, which are embedded into the constraint-based models of metabolism. Applications to single-cell sequencing data of glioblastoma brain tumors generate predictions about the effects of pharmaceutical interventions on the regulatory network and downstream metabolisms in different cell types from the tumor microenvironment. The model presents possible insights into treatments that could potentially suppress anaerobic metabolism in malignant cells with minimal impact on other cell types' metabolism. The proposed integrated model can guide therapeutic target prioritization, the formulation of combination therapies, and future drug discovery. This model integration framework is also generalizable to other applications, such as different cell types, organisms, and diseases.
Collapse
Affiliation(s)
- Tiange Shi
- University at Buffalo, Biostatistics, Buffalo, USA
| | - Han Yu
- Roswell Park Comprehensive Cancer Center, Biostatistics and Bioinformatics, Buffalo, USA
| | - Rachael Hageman Blair
- University at Buffalo, Biostatistics, Institute for Artificial Intelligence and Data Science, Buffalo, USA
| |
Collapse
|
12
|
Likonen D, Pinchasi M, Beery E, Sarsor Z, Signorini LF, Gervits A, Sharan R, Lahav M, Raanani P, Uziel O. Exosomal telomerase transcripts reprogram the microRNA transcriptome profile of fibroblasts and partially contribute to CAF formation. Sci Rep 2022; 12:16415. [PMID: 36180493 PMCID: PMC9525320 DOI: 10.1038/s41598-022-20186-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 09/09/2022] [Indexed: 11/25/2022] Open
Abstract
It is now well accepted that cancer cells change their microenvironment from normal to tumor-supportive state to provide sustained tumor growth, metastasis and drug resistance. These processes are partially carried out by exosomes, nano-sized vesicles secreted from cells, shuttled from donor to recipient cells containing a cargo of nucleic acids, proteins and lipids. By transferring biologically active molecules, cancer-derived exosomes may transform microenvironmental cells to become tumor supportive. Telomerase activity is regarded as a hallmark of cancer. We have recently shown that the transcript of human telomerase reverse transcriptase (hTERT), is packaged in cancer cells derived- exosomes. Following the engulfment of the hTERT transcript into fibroblasts, it is translated into a fully active enzyme [after assembly with its RNA component (hTERC) subunit]. Telomerase activity in the recipient, otherwise telomerase negative cells, provides them with a survival advantage. Here we show that exosomal telomerase might play a role in modifying normal fibroblasts into cancer associated fibroblasts (CAFs) by upregulating \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\mathrm{\alpha }$$\end{document}αSMA and Vimentin, two CAF markers. We also show that telomerase activity changes the transcriptome of microRNA in these fibroblasts. By ectopically expressing microRNA 342, one of the top identified microRNAs, we show that it may mediate the proliferative phenotype that these cells acquire upon taking-up exosomal hTERT, providing them with a survival advantage.
Collapse
Affiliation(s)
- Daniela Likonen
- The Felsenstein Medical Research Center, Petah-Tikva, Israel
| | - Maria Pinchasi
- The Felsenstein Medical Research Center, Petah-Tikva, Israel
| | - Einat Beery
- The Felsenstein Medical Research Center, Petah-Tikva, Israel
| | - Zinab Sarsor
- The Felsenstein Medical Research Center, Petah-Tikva, Israel
| | | | - Asia Gervits
- School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
| | - Roded Sharan
- School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
| | - Meir Lahav
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Pia Raanani
- The Felsenstein Medical Research Center, Petah-Tikva, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel
| | - Orit Uziel
- The Felsenstein Medical Research Center, Petah-Tikva, Israel. .,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel. .,Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel.
| |
Collapse
|
13
|
Shepherd TG, Dick FA. Principles of dormancy evident in high-grade serous ovarian cancer. Cell Div 2022; 17:2. [PMID: 35321751 PMCID: PMC8944075 DOI: 10.1186/s13008-022-00079-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/16/2022] [Indexed: 11/17/2022] Open
Abstract
In cancer, dormancy refers to a clinical state in which microscopic residual disease becomes non-proliferative and is largely refractory to chemotherapy. Dormancy was first described in breast cancer where disease can remain undetected for decades, ultimately leading to relapse and clinical presentation of the original malignancy. A long latency period can be explained by withdrawal from cell proliferation (cellular dormancy), or a balance between proliferation and cell death that retains low levels of residual disease (tumor mass dormancy). Research into cellular dormancy has revealed features that define this state. They include arrest of cell proliferation, altered cellular metabolism, and unique cell dependencies and interactions with the microenvironment. These characteristics can be shared by dormant cells derived from disparate primary disease sites, suggesting common features exist between them. High-grade serous ovarian cancer (HGSOC) disseminates to locations throughout the abdominal cavity by means of cellular aggregates called spheroids. These growth-arrested and therapy-resistant cells are a strong contributor to disease relapse. In this review, we discuss the similarities and differences between ovarian cancer cells in spheroids and dormant properties reported for other cancer disease sites. This reveals that elements of dormancy, such as cell cycle control mechanisms and changes to metabolism, may be similar across most forms of cellular dormancy. However, HGSOC-specific aspects of spheroid biology, including the extracellular matrix organization and microenvironment, are obligatorily disease site specific. Collectively, our critical review of current literature highlights places where HGSOC cell dormancy may offer a more tractable experimental approach to understand broad principles of cellular dormancy in cancer.
Collapse
Affiliation(s)
- Trevor G Shepherd
- London Regional Cancer Program, London Health Sciences Centre, London, ON, N6A 5W9, Canada.,Department of Obstetrics & Gynaecology, Western University, London, ON, N6A 5C1, Canada
| | - Frederick A Dick
- London Regional Cancer Program, London Health Sciences Centre, London, ON, N6A 5W9, Canada. .,Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 5C1, Canada. .,Children's Health Research Institute, London, ON, N6A 4V2, Canada.
| |
Collapse
|
14
|
Okikawa S, Morine Y, Saito Y, Yamada S, Tokuda K, Teraoku H, Miyazaki K, Yamashita S, Ikemoto T, Imura S, Shimada M. Inhibition of the VEGF signaling pathway attenuates tumor‑associated macrophage activity in liver cancer. Oncol Rep 2022; 47:71. [PMID: 35169858 PMCID: PMC8867251 DOI: 10.3892/or.2022.8282] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/13/2022] [Indexed: 11/06/2022] Open
Abstract
Tumor-associated macrophage (TAMs) are paramount for tumor progression and immune tolerance in the tumor microenvironment of various types of cancer, including liver cancer. The aim of the present study was to investigate the effect of vascular endothelial growth factor (VEGF) inhibition on TAM polarization and function during their interactions with macrophages and liver cancer cells. TAMs were induced by culturing M0 macrophages with cancer cell-conditioned medium. TAMs cultured with cancer cell-conditioned medium and vascular endothelial growth factor (VEGF) inhibitor were defined as modified TAMs, and the expression levels of TAM-associated markers and VEGF receptor 2 were evaluated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The effects of TAMs and modified TAMs on cancer cell proliferation and migration were investigated using conditioned medium. Programmed death-ligand 1 (PD-L1) mRNA expression in modified TAMs and cancer cells cultured in modified TAM-conditioned medium (TAM-CM) for 48 h was examined using RT-qPCR. In order to investigate signaling pathways in macrophages, western blot analysis was performed. CD163 and CD206 and M2 macrophage marker expression was upregulated in TAMs and modified TAMs. Modified TAM-CM exhibited a decreased ability to promote cancer cell proliferation and migration in comparison with the use of TAM-CM. The VEGF concentration was significantly higher in the TAMs than in M0 macrophages; however, the modified TAMs displayed a significantly lower VEGF secretion than TAMs. PD-L1 expression was decreased in modified TAMs as compared with TAMs. Western blot analysis revealed that the Akt/mTOR signaling pathway was significantly suppressed in the modified TAMs compared with TAMs. It was observed that TAMs cultured in a VEGF-depleted environment displayed lower secretion levels of cytokines involved in tumor progression and a decreased immune tolerance-inducing ability. On the whole, the results of the present study suggested that VEGF inhibition in TAMs may be a potential therapeutic target for liver cancer.
Collapse
Affiliation(s)
- Shohei Okikawa
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Yuji Morine
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Yu Saito
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Shinichiro Yamada
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Kazunori Tokuda
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Hiroki Teraoku
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Katsuki Miyazaki
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Shoko Yamashita
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Tetsuya Ikemoto
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Satoru Imura
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| | - Mitsuo Shimada
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8503, Japan
| |
Collapse
|
15
|
Beeghly GF, Amofa KY, Fischbach C, Kumar S. Regulation of Tumor Invasion by the Physical Microenvironment: Lessons from Breast and Brain Cancer. Annu Rev Biomed Eng 2022; 24:29-59. [PMID: 35119915 DOI: 10.1146/annurev-bioeng-110220-115419] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The success of anticancer therapies is often limited by heterogeneity within and between tumors. While much attention has been devoted to understanding the intrinsic molecular diversity of tumor cells, the surrounding tissue microenvironment is also highly complex and coevolves with tumor cells to drive clinical outcomes. Here, we propose that diverse types of solid tumors share common physical motifs that change in time and space, serving as universal regulators of malignancy. We use breast cancer and glioblastoma as instructive examples and highlight how invasion in both diseases is driven by the appropriation of structural guidance cues, contact-dependent heterotypic interactions with stromal cells, and elevated interstitial fluid pressure and flow. We discuss how engineering strategies show increasing value for measuring and modeling these physical properties for mechanistic studies. Moreover, engineered systems offer great promise for developing and testing novel therapies that improve patient prognosis by normalizing the physical tumor microenvironment. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Garrett F Beeghly
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA;
| | - Kwasi Y Amofa
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA; .,Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, USA
| | - Sanjay Kumar
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, USA.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
16
|
Lee H, Park SJ, Hong S, Lim SW, Kim S. Deletion of IP6K1 in mice accelerates tumor growth by dysregulating the tumor-immune microenvironment. Anim Cells Syst (Seoul) 2022; 26:19-27. [PMID: 35308129 PMCID: PMC8928833 DOI: 10.1080/19768354.2022.2029560] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A family of inositol hexakisphosphate kinases (IP6Ks) catalyzes the production of inositol pyrophosphate IP7 (5-diphosphoinositolpentakisphosphate) which is known to modulate various biological events such as cell growth. While targeting IP6K1 in various cancer cells has been well reported to control cancer cell motility and invasiveness, the role of host IP6K1 in tumor progression remains unknown. By using a syngeneic MC38 murine mouse colon carcinoma model, here we examined how host IP6K1 in the tumor microenvironment influences tumor growth. In IP6K1 knockout (KO) mice, the growth of MC38 tumor cells was markedly accelerated and host survival was significantly shortened compared with wild-type (WT). Our flow cytometric analysis revealed that tumors grown in IP6K1 KO mice had lower immune suppressive myeloid cells and M1 polarized macrophages. Notably, infiltration of both antigen-presenting dendritic cells and CD8+ cytotoxic T lymphocytes into the tumor tissues was remarkably abrogated in IP6K1 KO condition. These studies suggest that enhanced tumor growth in IP6K1 KO mice resulted from reduced anti-tumor immunity due to disturbed immune cell actions in the tumor microenvironment. In conclusion, we demonstrate that host IP6K1 acts as a tumor suppressor, most likely by fine-tuning diverse tumor-immune cell interactions, which might have implications for improving the host response against cancer progression.
Collapse
Affiliation(s)
- Haein Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seung Ju Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Sehoon Hong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seol-Wa Lim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seyun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- KAIST Institute for the BioCentury, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
17
|
Seong JB, Kim B, Kim S, Kim MH, Park YH, Lee Y, Lee HJ, Hong CW, Lee DS. Macrophage peroxiredoxin 5 deficiency promotes lung cancer progression via ROS-dependent M2-like polarization. Free Radic Biol Med 2021; 176:322-334. [PMID: 34637923 DOI: 10.1016/j.freeradbiomed.2021.10.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/24/2022]
Abstract
Strategies for cancer treatment have traditionally focused on suppressing cancer cell behavior, but many recent studies have demonstrated that regulating the tumor microenvironment (TME) can also inhibit disease progression. Macrophages are major TME components, and the direction of phenotype polarization is known to regulate tumor behavior, with M2-like polarization promoting progression. It is also known that reactive oxygen species (ROS) in macrophages drive M2 polarization, and M2 polarization promote lung cancer progression. Lung cancer patients with lower expression of the antioxidant enzyme peroxiredoxin 5 (Prx5) demonstrate poorer survival. This study revealed that Prx5 deficiency in macrophages induced M2 macrophage polarization by lung cancer. We report that injection of lung cancer cells produced larger tumors in Prx5-deficit mice than wild-type mice independent of cancer cell Prx5 expression. Through co-culture with lung cancer cell lines, Prx5-deficient macrophages exhibited M2 polarization, and reduced expression levels of the M1-associated inflammatory factors iNOS, TNFα, and Il-1β. Moreover, these Prx5-deficient macrophages promoted the proliferation and migration of co-cultured lung cancer cells. Conversely, suppression of ROS generation by N-acetyl cysteine (NAC) inhibited the M2-like polarization of Prx5-deficient macrophages, increased expression levels of inflammatory factors, inhibited the proliferation and migration of co-cultured lung cancer cells, and suppressed tumor growth in mice. These findings suggest that blocking the M2 polarization of macrophages may promote lung cancer regression.
Collapse
Affiliation(s)
- Jung Bae Seong
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea; National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Bokyung Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Soyoon Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Mi Hye Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Young-Ho Park
- Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Hong Jun Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea; Research Institute eBiogen Inc., Seoul, Republic of Korea
| | - Chang-Won Hong
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
18
|
LncRNAs in tumor microenvironment: The potential target for cancer treatment with natural compounds and chemical drugs. Biochem Pharmacol 2021; 193:114802. [PMID: 34678226 DOI: 10.1016/j.bcp.2021.114802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022]
Abstract
It was thought that originally long non-coding RNAs (lncRNAs) were a kind of RNAs without any encoding function. Recently, a variety of studies have shown that lncRNAs play important roles in many life activities. The abnormal expression of lncRNAs in tumor microenvironment (TME) usually promotes the proliferation, migration, and drug resistance of tumor cells through direct or indirect effects, which also usually predicts the poor prognosis. The regulation of lncRNAs expression in TME could significantly inhibit tumor progress. However, the interaction between lncRNAs and TME has not been fully defined at present. Therefore, this paper provided the systemic summary of their interaction and natural products and chemicals targeting lncRNAs in cancer treatment. Currently, the strategies of cancer treatment still have their limits. Understanding the relationship between TME and lncRNAs can help us to realize breakthrough strategy for tumor treatment.
Collapse
|
19
|
Combinatorial therapy in tumor microenvironment: Where do we stand? Biochim Biophys Acta Rev Cancer 2021; 1876:188585. [PMID: 34224836 DOI: 10.1016/j.bbcan.2021.188585] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/28/2021] [Accepted: 06/23/2021] [Indexed: 01/09/2023]
Abstract
The tumor microenvironment plays a pivotal role in tumor initiation and progression by creating a dynamic interaction with cancer cells. The tumor microenvironment consists of various cellular components, including endothelial cells, fibroblasts, pericytes, adipocytes, immune cells, cancer stem cells and vasculature, which provide a sustained environment for cancer cell proliferation. Currently, targeting tumor microenvironment is increasingly being explored as a novel approach to improve cancer therapeutics, as it influences the growth and expansion of malignant cells in various ways. Despite continuous advancements in targeted therapies for cancer treatment, drug resistance, toxicity and immune escape mechanisms are the basis of treatment failure and cancer escape. Targeting tumor microenvironment efficiently with approved drugs and combination therapy is the solution to this enduring challenge that involves combining more than one treatment modality such as chemotherapy, surgery, radiotherapy, immunotherapy and nanotherapy that can effectively and synergistically target the critical pathways associated with disease pathogenesis. This review shed light on the composition of the tumor microenvironment, interaction of different components within tumor microenvironment with tumor cells and associated hallmarks, the current status of combinatorial therapies being developed, and various growing advancements. Furthermore, computational tools can also be used to monitor the significance and outcome of therapies being developed. We addressed the perceived barriers and regulatory hurdles in developing a combinatorial regimen and evaluated the present status of these therapies in the clinic. The accumulating depth of knowledge about the tumor microenvironment in cancer may facilitate further development of effective treatment modalities. This review presents the tumor microenvironment as a sweeping landscape for developing novel cancer therapies.
Collapse
|
20
|
Wang L, Sun X, Jin C, Fan Y, Xue F. Identification of Tumor Microenvironment-Related Prognostic Biomarkers for Ovarian Serous Cancer 3-Year Mortality Using Targeted Maximum Likelihood Estimation: A TCGA Data Mining Study. Front Genet 2021; 12:625145. [PMID: 34149794 PMCID: PMC8211425 DOI: 10.3389/fgene.2021.625145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/11/2021] [Indexed: 02/01/2023] Open
Abstract
Ovarian serous cancer (OSC) is one of the leading causes of death across the world. The role of the tumor microenvironment (TME) in OSC has received increasing attention. Targeted maximum likelihood estimation (TMLE) is developed under a counterfactual framework to produce effect estimation for both the population level and individual level. In this study, we aim to identify TME-related genes and using the TMLE method to estimate their effects on the 3-year mortality of OSC. In total, 285 OSC patients from the TCGA database constituted the studying population. ESTIMATE algorithm was implemented to evaluate immune and stromal components in TME. Differential analysis between high-score and low-score groups regarding ImmuneScore and StromalScore was performed to select shared differential expressed genes (DEGs). Univariate logistic regression analysis was followed to evaluate associations between DEGs and clinical pathologic factors with 3-year mortality. TMLE analysis was conducted to estimate the average effect (AE), individual effect (IE), and marginal odds ratio (MOR). The validation was performed using three datasets from Gene Expression Omnibus (GEO) database. Additionally, 355 DEGs were selected after differential analysis, and 12 genes from DEGs were significant after univariate logistic regression. Four genes remained significant after TMLE analysis. In specific, ARID3C and FREM2 were negatively correlated with OSC 3-year mortality. CROCC2 and PTF1A were positively correlated with OSC 3-year mortality. Combining of ESTIMATE algorithm and TMLE algorithm, we identified four TME-related genes in OSC. AEs were estimated to provide averaged effects based on the population level, while IEs were estimated to provide individualized effects and may be helpful for precision medicine.
Collapse
Affiliation(s)
- Lu Wang
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoru Sun
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuandi Jin
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Fan
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Xi’an Jiaotong University, Xi’an, China
| | - Fuzhong Xue
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
21
|
Petrou A, Fesatidou M, Geronikaki A. Thiazole Ring-A Biologically Active Scaffold. Molecules 2021; 26:3166. [PMID: 34070661 PMCID: PMC8198555 DOI: 10.3390/molecules26113166] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/15/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Thiazole is a good pharmacophore nucleus due to its various pharmaceutical applications. Its derivatives have a wide range of biological activities such as antioxidant, analgesic, and antimicrobial including antibacterial, antifungal, antimalarial, anticancer, antiallergic, antihypertensive, anti-inflammatory, and antipsychotic. Indeed, the thiazole scaffold is contained in more than 18 FDA-approved drugs as well as in numerous experimental drugs. OBJECTIVE To summarize recent literature on the biological activities of thiazole ring-containing compounds Methods: A literature survey regarding the topics from the year 2015 up to now was carried out. Older publications were not included, since they were previously analyzed in available peer reviews. RESULTS Nearly 124 research articles were found, critically analyzed, and arranged regarding the synthesis and biological activities of thiazoles derivatives in the last 5 years.
Collapse
Affiliation(s)
| | | | - Athina Geronikaki
- School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.P.); (M.F.)
| |
Collapse
|
22
|
Yang B, Xie J, Li Z, Su D, Lin L, Guo X, Fu Z, Zhou Q, Lu Y. Seven-gene signature on tumor microenvironment for predicting the prognosis of patients with pancreatic cancer. Gland Surg 2021; 10:1397-1409. [PMID: 33968691 DOI: 10.21037/gs-21-28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background The aim of the present study was to construct a novel gene signature on the tumor microenvironment (TME) to predict the prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). Methods We downloaded gene expression profiles and clinical information of PDAC from The Cancer Genome Atlas (TCGA) datasets, as well as Gene Expression Omnibus (GEO) datasets (GSE78229, GSE62452, and GSE28735). Differentially expressed genes were generated by comparing high versus low score groups of immune/stromal subgroups based on the Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data algorithm. Subsequently, a prognostic risk score model was constructed and validated through univariate and multivariate Cox regression analyses. Finally, functional enrichment analysis and protein-protein interactions were performed to predict the functional implication of the prognostic model. Results We picked out 1,797 upregulated genes in immune groups and stromal groups. Through further analysis, we constructed a 7-gene signature on the TME. The risk score from the model effectively differentiated patients into high-risk and low-risk groups with different overall survival and was validated by GEO datasets. A functional analysis suggested that 7 selected genes and their co-expressed genes were mainly enriched in immune response, extracellular structure organization, and cell adhesion molecule binding. Conclusions Our results showed that the 7-gene model on the TME can be used to assess the prognosis of patients with PDAC.
Collapse
Affiliation(s)
- Bin Yang
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinghua Xie
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiguo Li
- Department of Thoracic Surgery, the Second People Hospital of Foshan, Foshan, China
| | - Dan Su
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Longfa Lin
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofeng Guo
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiqiang Fu
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Quanbo Zhou
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanan Lu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Lu Y, Hou K, Li M, Wu X, Yuan S. Exosome-Delivered LncHEIH Promotes Gastric Cancer Progression by Upregulating EZH2 and Stimulating Methylation of the GSDME Promoter. Front Cell Dev Biol 2020; 8:571297. [PMID: 33163491 PMCID: PMC7591465 DOI: 10.3389/fcell.2020.571297] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/22/2020] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer is the third leading cause of cancer-related deaths worldwide and is characterized by poor survival and high recurrence rates. Long non-coding RNAs (lncRNAs) have gained considerable attention in recent years as prognostic markers and gene regulators in various cancers. Here, we found that lncHEIH was upregulated in gastric cancer tissues and cell lines and positively correlated with high expression levels of EZH2. Mechanistically, the lncHEIH-EZH2 axis could promote the progression of gastric cancer. In addition, lncHEIH encapsulated in exosomes was released by gastric cancer cells and then absorbed by normal gastric cells. The uptake of lncHEIH resulted in the upregulation of EZH2, which inhibited the expression of the tumor suppressor GSDME by methylation of the GSDME promoter, promoting the malignant transformation of normal gastric cells. Overall, lncHEIH promotes gastric cancer progression by upregulating the expression of EZH2 and reducing the expression of GSDME in normal cells to induce malignant cell proliferation and migration, indicating its potential as a target in gastric cancer therapy.
Collapse
Affiliation(s)
- Yan Lu
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, Key Laboratory of Gene Engineering of the Ministry of Education, College of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Department of Gastrointestinal Surgery, The 8th Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou, China
| | - Kaiqing Hou
- Department of Gastrointestinal Surgery, Haikou City People’s Hospital, Haikou, China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou, China
| | - Xiaobin Wu
- Department of Gastrointestinal Surgery, The 8th Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shaochun Yuan
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, Key Laboratory of Gene Engineering of the Ministry of Education, College of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
24
|
Nezhadi S, Saadat E, Handali S, Dorkoosh F. Nanomedicine and chemotherapeutics drug delivery: challenges and opportunities. J Drug Target 2020; 29:185-198. [PMID: 32772739 DOI: 10.1080/1061186x.2020.1808000] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer is considered as one of the biggest threats to humans worldwide. Researchers suggest that tumour is not just a single mass, it comprises cancerous cells surrounded by noncancerous cells such as immune cells, adipocytes and cancer stem cells (CSCs) in the extracellular matrix (ECM) containing distinct components such as proteins, glycoproteins and enzymes; thus tumour microenvironment (TME) is partially complex. Multiple interactions happen in the dynamic microenvironment (ME) lead to an acidic, hypoxic and stiff ME that is considered as one of the major contributors to cancer progression and metastasis. Furthermore, TME involves in drug resistance mechanisms and affects enhanced permeability and retention (EPR) in tumours. In such a scenario, the first step to accomplish satisfying results is the identification and recognition of this ME. Then designing proper drug delivery systems can perform selectively towards cancerous cells. In this way, several targeting and stimuli/enzyme responsive drug delivery systems have been designed. More importantly, it is necessary to design a drug delivery system that can penetrate deeper into the tumours, efficiently and selectively. Various drug delivery systems such as exosomes and size-switchable nanocarriers (NCs) could decrease side effects and increase tumour treatment results by selective accumulation in tumours. In this review, TME features, current drug delivery approaches, challenges and promising strategies towards cancer treatment are discussed.
Collapse
Affiliation(s)
- Sepideh Nezhadi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Ir an
| | | | - Somayeh Handali
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Ir an.,Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
The not-so-sweet side of sugar: Influence of the microenvironment on the processes that unleash cancer. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165960. [PMID: 32919034 DOI: 10.1016/j.bbadis.2020.165960] [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: 05/21/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022]
Abstract
The role of "aerobic glycolysis" in cancer has been examined often in the past. Results from those studies, most of which were performed on two dimensional conditions (2D, tissue culture plastic), demonstrate that aerobic glycolysis occurs as a consequence of oncogenic events. These oncogenic events often drive malignant cell growth and survival. Although 2D based experiments are useful in elucidating the molecular mechanisms of oncogenesis, they fail to take contributions of the extracellular microenvironment into account. Indeed we, and others, have shown that the cellular microenvironment is essential in regulating processes that induce and/or suppress the malignant phenotype/properties. This regulation between the cell and its microenvironment is both dynamic and reciprocal and involves the integration of cellular signaling networks in the right context. Therefore, given our previous demonstration of the effect of the microenvironment including tissue architecture and media composition on gene expression and the integration of signaling events observed in three-dimension (3D), we hypothesized that glucose uptake and metabolism must also be essential components of the tissue's signal "integration plan" - that is, if uptake and metabolism of glucose were hyperactivated, the canonical oncogenic pathways should also be similarly activated. This hypothesis, if proven true, suggests that direct inhibition of glucose metabolism in cancer cells should either suppress or revert the malignant phenotype in 3D. Here, we review the up-to-date progress that has been made towards understanding the role that glucose metabolism plays in oncogenesis and re-establishing basally polarized acini in malignant human breast cells.
Collapse
|
26
|
Collagen and fibronectin promote an aggressive cancer phenotype in breast cancer cells but drive autonomous gene expression patterns. Gene 2020; 761:145024. [PMID: 32755659 DOI: 10.1016/j.gene.2020.145024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/08/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023]
Abstract
Understanding how various pathologies of breast cancer respond to their environment may be imperative in the creation of novel therapeutic targets. Central to the organisation and behaviour of cells within the tumour microenvironment is the extracellular matrix (ECM), a meshwork of fibrous proteins and glycoproteins that directly influences cell behaviour and the bioavailability of signalling molecules. Our appreciation on how the composition of the ECM can influence cancer behaviour has evolved significantly and although we are highly cognisant of the dramatic impact the ECM can have on cancer cell behaviour, we continue to neglect this during diagnosis and treatment. In the following study, we aimed to identify how three breast cancer cell lines respond functionally and genetically to common components of the ECM. Using real time and end point assays we have identified similar patterns of behaviour among the three breast cancer cell lines in response to commonly found ECM components of the breast. Using a selected gene panel, we have been able to identify cell line specific changes in gene differentiation when breast cancer cells are in contact with these elements. Although the response of our cells to these elements differ at the genetic level, their functional responses are consistent. This work adds to the growing arguments that highlight a need for histologically assessing ECM composition of breast tumours. In particular monitoring of fibrous protein deposition at the site of malignancy could provide critical information during clinical assessment influencing disease prognosis and treatment decisions for breast cancer patients.
Collapse
|
27
|
Leask A. A centralized communication network: Recent insights into the role of the cancer associated fibroblast in the development of drug resistance in tumors. Semin Cell Dev Biol 2020; 101:111-114. [DOI: 10.1016/j.semcdb.2019.10.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022]
|
28
|
Mavrogonatou E, Pratsinis H, Kletsas D. The role of senescence in cancer development. Semin Cancer Biol 2020; 62:182-191. [DOI: 10.1016/j.semcancer.2019.06.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 02/07/2023]
|
29
|
Caveolin-1 regulates the ASMase/ceramide-mediated radiation response of endothelial cells in the context of tumor-stroma interactions. Cell Death Dis 2020; 11:228. [PMID: 32273493 PMCID: PMC7145831 DOI: 10.1038/s41419-020-2418-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/21/2022]
Abstract
The integral membrane protein caveolin-1 (CAV1) plays a central role in radioresistance-mediating tumor–stroma interactions of advanced prostate cancer (PCa). Among the tumor–stroma, endothelial cells (EC) evolved as critical determinants of the radiation response. CAV1 deficiency in angiogenic EC was already shown to account for increased apoptosis rates of irradiated EC. This study explores the potential impact of differential CAV1 levels in EC on the acid sphingomyelinase (ASMase)/ceramide pathway as a key player in the regulation of EC apoptosis upon irradiation and cancer cell radioresistance. Enhanced apoptosis sensitivity of CAV1-deficient EC was associated with increased ASMase activity, ceramide generation, formation of large lipid platforms, and finally an altered p38 mitogen-activated protein kinase (MAPK)/heat-shock protein 27 (HSP27)/AKT (protein kinase B, PKB) signaling. CAV1-deficient EC increased the growth delay of LNCaP and PC3 PCa cells upon radiation treatment in direct 3D spheroid co-cultures. Exogenous C6 and C16 ceramide treatment in parallel increased the growth delay of PCa spheroids and induced PCa cell apoptosis. Analysis of the respective ceramide species in PCa cells with increased CAV1 levels like those typically found in radio-resistant advanced prostate tumors further revealed an upregulation of unsaturated C24:1 ceramide that might scavenge the effects of EC-derived apoptosis-inducing C16 ceramide. Higher ASMase as well as ceramide levels could be confirmed by immunohistochemistry in human advanced prostate cancer specimen bearing characteristic CAV1 tumor–stroma alterations. Conclusively, CAV1 critically regulates the generation of ceramide-dependent (re-)organization of the plasma membrane that in turn affects the radiation response of EC and adjacent PCa cells. Understanding the CAV1-dependent crosstalk between tumor cells and the host-derived tumor microvasculature and its impact on radiosensitivity may allow to define a rational strategy for overcoming tumor radiation resistance improving clinical outcomes by targeting CAV1.
Collapse
|
30
|
Baghban R, Roshangar L, Jahanban-Esfahlan R, Seidi K, Ebrahimi-Kalan A, Jaymand M, Kolahian S, Javaheri T, Zare P. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal 2020; 18:59. [PMID: 32264958 PMCID: PMC7140346 DOI: 10.1186/s12964-020-0530-4] [Citation(s) in RCA: 888] [Impact Index Per Article: 222.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/05/2020] [Indexed: 02/07/2023] Open
Abstract
The dynamic interactions of cancer cells with their microenvironment consisting of stromal cells (cellular part) and extracellular matrix (ECM) components (non-cellular) is essential to stimulate the heterogeneity of cancer cell, clonal evolution and to increase the multidrug resistance ending in cancer cell progression and metastasis. The reciprocal cell-cell/ECM interaction and tumor cell hijacking of non-malignant cells force stromal cells to lose their function and acquire new phenotypes that promote development and invasion of tumor cells. Understanding the underlying cellular and molecular mechanisms governing these interactions can be used as a novel strategy to indirectly disrupt cancer cell interplay and contribute to the development of efficient and safe therapeutic strategies to fight cancer. Furthermore, the tumor-derived circulating materials can also be used as cancer diagnostic tools to precisely predict and monitor the outcome of therapy. This review evaluates such potentials in various advanced cancer models, with a focus on 3D systems as well as lab-on-chip devices. Video abstract.
Collapse
Affiliation(s)
- Roghayyeh Baghban
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khaled Seidi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committees, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Department of Neurosciences and Cognitive, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saeed Kolahian
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, University Hospital Tuebingen, Tuebingen, Germany
| | - Tahereh Javaheri
- Health Informatics Lab, Metropolitan College, Boston University, Boston, USA
| | - Peyman Zare
- Dioscuri Center of Chromatin Biology and Epigenomics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Faculty of Medicine, Cardinal Stefan Wyszyński University in Warsaw, 01-938 Warsaw, Poland
| |
Collapse
|
31
|
Tunneling Nanotubes and Tumor Microtubes in Cancer. Cancers (Basel) 2020; 12:cancers12040857. [PMID: 32244839 PMCID: PMC7226329 DOI: 10.3390/cancers12040857] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
Intercellular communication among cancer cells and their microenvironment is crucial to disease progression. The mechanisms by which communication occurs between distant cells in a tumor matrix remain poorly understood. In the last two decades, experimental evidence from different groups proved the existence of thin membranous tubes that interconnect cells, named tunneling nanotubes, tumor microtubes, cytonemes or membrane bridges. These highly dynamic membrane protrusions are conduits for direct cell-to-cell communication, particularly for intercellular signaling and transport of cellular cargo over long distances. Tunneling nanotubes and tumor microtubes may play an important role in the pathogenesis of cancer. They may contribute to the resistance of tumor cells against treatments such as surgery, radio- and chemotherapy. In this review, we present the current knowledge about the structure and function of tunneling nanotubes and tumor microtubes in cancer and discuss the therapeutic potential of membrane tubes in cancer treatment.
Collapse
|
32
|
Xun Y, Yang H, Li J, Wu F, Liu F. CXC Chemokine Receptors in the Tumor Microenvironment and an Update of Antagonist Development. Rev Physiol Biochem Pharmacol 2020; 178:1-40. [PMID: 32816229 DOI: 10.1007/112_2020_35] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chemokine receptors, a diverse group within the seven-transmembrane G protein-coupled receptor superfamily, are frequently overexpressed in malignant tumors. Ligand binding activates multiple downstream signal transduction cascades that drive tumor growth and metastasis, resulting in poor clinical outcome. These receptors are thus considered promising targets for anti-tumor therapy. This article reviews recent studies on the expression and function of CXC chemokine receptors in various tumor microenvironments and recent developments in cancer therapy using CXC chemokine receptor antagonists.
Collapse
Affiliation(s)
- Yang Xun
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Hua Yang
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Jiekai Li
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Fuling Wu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Fang Liu
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China.
| |
Collapse
|
33
|
Targeting the Tumor Microenvironment: An Unexplored Strategy for Mutant KRAS Tumors. Cancers (Basel) 2019; 11:cancers11122010. [PMID: 31847096 PMCID: PMC6966533 DOI: 10.3390/cancers11122010] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/09/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022] Open
Abstract
Current evidence strongly suggests that cancer cells depend on the microenvironment in order to thrive. In fact, signals from the surrounding tumor microenvironment are crucial for cancer cells´ aggressiveness, altering their expression profile and favoring their metastatic potential. As such, targeting the tumor microenvironment to impair cancer progression became an attractive therapeutic option. Interestingly, it has been shown that oncogenic KRAS signaling promotes a pro-tumorigenic microenvironment, and the associated crosstalk alters the expression profile of cancer cells. These findings award KRAS a key role in controlling the interactions between cancer cells and the microenvironment, granting cancer a poor prognosis. Given the lack of effective approaches to target KRAS itself or its downstream effectors in the clinic, exploring such interactions may open new perspectives on possible therapeutic strategies to hinder mutant KRAS tumors. This review highlights those communications and their implications for the development of effective therapies or to provide insights regarding response to existing regimens.
Collapse
|
34
|
Bellefeuille SD, Molle CM, Gendron FP. Reviewing the role of P2Y receptors in specific gastrointestinal cancers. Purinergic Signal 2019; 15:451-463. [PMID: 31478181 PMCID: PMC6923304 DOI: 10.1007/s11302-019-09678-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular nucleotides are important intercellular signaling molecules that were found enriched in the tumor microenvironment. In fact, interfering with G protein-coupled P2Y receptor signaling has emerged as a promising therapeutic alternative to treat aggressive and difficult-to-manage cancers such as those affecting the gastrointestinal system. In this review, we will discuss the functions of P2Y receptors in gastrointestinal cancers with an emphasis on colorectal, hepatic, and pancreatic cancers. We will show that P2Y2 receptor up-regulation increases cancer cell proliferation, tumor growth, and metastasis in almost all studied gastrointestinal cancers. In contrast, we will present P2Y6 receptor as having opposing roles in colorectal cancer vs. gastric cancer. In colorectal cancer, the P2Y6 receptor induces carcinogenesis by inhibiting apoptosis, whereas P2Y6 suppresses gastric cancer tumor growth by reducing β-catenin transcriptional activity. The contribution of the P2Y11 receptor in the migration of liver and pancreatic cancer cells will be compared to its normal inhibitory function on this cellular process in ciliated cholangiocytes. Hence, we will demonstrate that the selective inhibition of the P2Y12 receptor activity in platelets was associated to a reduction in the risk of developing colorectal cancer and metastasis formation. We will succinctly review the role of P2Y1, P2Y4, P2Y13, and P2Y14 receptors as the knowledge for these receptors in gastrointestinal cancers is sparse. Finally, redundant ligand selectivity, nucleotide high lability, cell context, and antibody reliability will be presented as the main difficulties in defining P2Y receptor functions in gastrointestinal cancers.
Collapse
Affiliation(s)
- Steve Dagenais Bellefeuille
- Département d’anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, 3201 rue Jean-Mignault, Sherbrooke, Québec, J1E 4K8 Canada
| | - Caroline M. Molle
- Département d’anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, 3201 rue Jean-Mignault, Sherbrooke, Québec, J1E 4K8 Canada
| | - Fernand-Pierre Gendron
- Département d’anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, 3201 rue Jean-Mignault, Sherbrooke, Québec, J1E 4K8 Canada
| |
Collapse
|
35
|
Houghton PJ, Kurmasheva RT. Challenges and Opportunities for Childhood Cancer Drug Development. Pharmacol Rev 2019; 71:671-697. [PMID: 31558580 PMCID: PMC6768308 DOI: 10.1124/pr.118.016972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer in children is rare with approximately 15,700 new cases diagnosed in the United States annually. Through use of multimodality therapy (surgery, radiation therapy, and aggressive chemotherapy), 70% of patients will be "cured" of their disease, and 5-year event-free survival exceeds 80%. However, for patients surviving their malignancy, therapy-related long-term adverse effects are severe, with an estimated 50% having chronic life-threatening toxicities related to therapy in their fourth or fifth decade of life. While overall intensive therapy with cytotoxic agents continues to reduce cancer-related mortality, new understanding of the molecular etiology of many childhood cancers offers an opportunity to redirect efforts to develop effective, less genotoxic therapeutic options, including agents that target oncogenic drivers directly, and the potential for use of agents that target the tumor microenvironment and immune-directed therapies. However, for many high-risk cancers, significant challenges remain.
Collapse
Affiliation(s)
- Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| |
Collapse
|
36
|
Dong Z, Yang Z, Hao Y, Feng L. Fabrication of H 2O 2-driven nanoreactors for innovative cancer treatments. NANOSCALE 2019; 11:16164-16186. [PMID: 31453999 DOI: 10.1039/c9nr04418c] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The increased production of hydrogen peroxide (H2O2) is a typical feature of cancerous cells. This feature is closely associated with elevated oxidative stress inside solid tumour microenvironments, which thus impairs either the growth of cancer cells or their sensitivity to many cancer therapeutics. To date, numerous innovative strategies that target tumour H2O2 have been designed for effective cancer treatment. More recently, with the rapid advancement of nanomedicine, several nanoreactors, which are highly efficient in converting endogenous H2O2 to more toxic reactive oxygen species, promoting in situ H2O2, or decomposing endogenous H2O2 to molecular oxygen for tumour hypoxia attenuation, have been designed and attempted for effective cancer treatment. This review focuses on the latest progress of such innovative H2O2-driven nanoreactor-mediated cancer treatments. Afterwards, future perspectives on the development of tumour H2O2-driven nanoreactor-mediated cancer treatments and their potential clinical translations will be discussed.
Collapse
Affiliation(s)
- Ziliang Dong
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Zhijuan Yang
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Yu Hao
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Liangzhu Feng
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
37
|
Roberts-Thomson SJ, Chalmers SB, Monteith GR. The Calcium-Signaling Toolkit in Cancer: Remodeling and Targeting. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035204. [PMID: 31088826 DOI: 10.1101/cshperspect.a035204] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Processes that are important in cancer progression, such as sustained cell growth, invasion to other organs, and resistance to cell death inducers, have a clear overlap with pathways regulated by Ca2+ signaling. It is therefore not surprising that proteins important in Ca2+ signaling, sometimes referred to as the "Ca2+ signaling toolkit," can contribute to cancer cell proliferation and invasiveness, and the ability of agents to induce cancer cell death. Ca2+ signaling is also critical in other aspects of cancer progression, including events in the tumor microenvironment and processes involved in the acquisition of resistance to anticancer therapies. This review will consider the role of Ca2+ signaling in tumor progression and highlight areas in which a better understanding of the interplay between the Ca2+-signaling toolkit and tumorigenesis is still required.
Collapse
Affiliation(s)
| | - Silke B Chalmers
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Gregory R Monteith
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia.,Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland 4072, Australia
| |
Collapse
|
38
|
Jahchan NS, Mujal AM, Pollack JL, Binnewies M, Sriram V, Reyno L, Krummel MF. Tuning the Tumor Myeloid Microenvironment to Fight Cancer. Front Immunol 2019; 10:1611. [PMID: 31402908 PMCID: PMC6673698 DOI: 10.3389/fimmu.2019.01611] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment (TME) of diverse cancer types is often characterized by high levels of infiltrating myeloid cells including monocytes, macrophages, dendritic cells, and granulocytes. These cells perform a variety of functions in the TME, varying from immune suppressive to immune stimulatory roles. In this review, we summarize the different myeloid cell populations in the TME and the intratumoral myeloid targeting approaches that are being clinically investigated, and discuss strategies that identify new myeloid subpopulations within the TME. The TME therapies include agents that modulate the functional activities of myeloid populations, that impact recruitment and survival of myeloid subpopulations, and that functionally reprogram or activate myeloid populations. We discuss the benefits, limitations and potential side effects of these therapeutic approaches.
Collapse
Affiliation(s)
| | - Adriana M. Mujal
- Department of Pathology, University of California, San Francisco, San Francisco, CA, United States
| | | | | | | | - Leonard Reyno
- Pionyr Immunotherapeutics, South San Francisco, CA, United States
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA, United States
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
39
|
Camargo S, Shamis Y, Assis A, Mitrani E. An in vivo Like Micro-Carcinoma Model. Front Oncol 2019; 9:410. [PMID: 31192122 PMCID: PMC6540606 DOI: 10.3389/fonc.2019.00410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/01/2019] [Indexed: 11/13/2022] Open
Abstract
We here present a novel micro-system which allows to reconstitute an in vivo lung carcinoma where the various constituting epithelial and/or stromal structural and/or cellular components can be incorporated at will. In contrast to various "organs on a chip" the model is based on the observation that in nature, epithelial cells are always supported by a connective tissue or stroma. The model is based on acellular micro-scaffolds of microscopic dimensions which enable seeded cells to obtain gases and nutrients through diffusion thus avoiding the need for vascularization. As a proof of concept, we show that in this model, Calu-3 cells can form a well-organized, continuous, polarized, one-layer epithelium lining the stromal derived alveolar cavities, and express a different pattern of tumor-related genes than when grown as standard monolayer cultures on plastic culture dishes. To our knowledge, this model, introduces for the first time a system where the function of carcinogenic cells can be tested in vitro in an environment that closely mimics the natural in vivo situation.
Collapse
Affiliation(s)
- Sandra Camargo
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yulia Shamis
- Department of Developmental and Regenerative Biology, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Assaf Assis
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eduardo Mitrani
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
40
|
Zhuang X, Zhang H, Hu G. Cancer and Microenvironment Plasticity: Double-Edged Swords in Metastasis. Trends Pharmacol Sci 2019; 40:419-429. [PMID: 31078320 DOI: 10.1016/j.tips.2019.04.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/07/2019] [Accepted: 04/12/2019] [Indexed: 02/07/2023]
Abstract
Cancer initiates at one site (primary tumor) and, in most cases, spreads to other distant organs (metastasis). During the multistep process of metastasis, primary tumor cells acquire cellular and phenotypic plasticity to survive and thrive in different environments. Moreover, cancer cells also utilize and educate microenvironmental components by reshaping them into accomplices of metastasis. Recent studies have identified a plethora of new molecular and cellular modulators of metastasis that have dynamic or even opposite roles, dominating the phenotypic plasticity of both tumoral and microenvironmental components. In this review we discuss their bipotential functions and the possible underpinning mechanisms, as well as their implications for targeted cancer therapy.
Collapse
Affiliation(s)
- Xueqian Zhuang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hao Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
41
|
Tang Y, Cui Y, Zhang S, Zhang L. The sensitivity and specificity of serum glycan-based biomarkers for cancer detection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 162:121-140. [PMID: 30905445 DOI: 10.1016/bs.pmbts.2019.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Most of clinically used serum biomarkers for cancer detection were established in early 1980s when the Nobel Prize in physiology or medicine was awarded for the "discovery of the principle for the production of monoclonal antibodies." Using this "Nobel" technology, various monoclonal antibodies were obtained when different types of cancer cells were injected into mice and the ligands on the cancer cell surface were characterized. Both aberrant glycan structures and aberrant glycan-associated glycoproteins were revealed as a common feature of cancer cell surfaces through the specific interactions with the monoclonal antibodies. These results indicate that the biosynthesis of the environment-sensitive glycan structures goes awry in cancer cells, which is beyond genetic mutations. Later on, the glycan-related biomarkers were detected in the sera of cancer patients and then developed into serum biomarkers, such as CA125, CA153, CA195, CA199, CA242, CA27.29, CA50, and CA724, which are still in clinical use as of today. During the past 30 years, even with the advancement of different OMICS technologies not limited to genomics, epigenomics, proteomics, glycomics, lipidomics, and metabolomics, very few serum biomarkers have been introduced into clinical practice. The reason is that most of the newly discovered cancer biomarkers are inferior in terms of sensitivity and specificity to these biomarkers. We will summarize the reported sensitivity and specificity of currently used cancer biomarkers, especially the glycan-related biomarkers, in the forms of tables and radar plots and discuss the pros and cons of currently used cancer biomarkers.
Collapse
Affiliation(s)
- Yang Tang
- Systems Biology and Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, China; School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Yidi Cui
- Systems Biology and Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, China; School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shufeng Zhang
- College of Chemistry, Tianjin Normal University, Tianjin, China
| | - Lijuan Zhang
- Systems Biology and Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, China; School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.
| |
Collapse
|
42
|
Tumor-Associated Macrophages Induce Endocrine Therapy Resistance in ER+ Breast Cancer Cells. Cancers (Basel) 2019; 11:cancers11020189. [PMID: 30736340 PMCID: PMC6406935 DOI: 10.3390/cancers11020189] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 02/07/2023] Open
Abstract
Antiestrogenic adjuvant treatments are first-line therapies in patients with breast cancer positive for estrogen receptor (ER+). Improvement of their treatment strategies is needed because most patients eventually acquire endocrine resistance and many others are initially refractory to anti-estrogen treatments. The tumor microenvironment plays essential roles in cancer development and progress; however, the molecular mechanisms underlying such effects remain poorly understood. Breast cancer cell lines co-cultured with TNF-α-conditioned macrophages were used as pro-inflammatory tumor microenvironment models. Proliferation, migration, and colony formation assays were performed to evaluate tamoxifen and ICI 182,780 resistance and confirmed in a mouse-xenograft model. Molecular mechanisms were investigated using cytokine antibody arrays, WB, ELISA, ChIP, siRNA, and qPCR-assays. In our simulated pro-inflammatory tumor microenvironment, tumor-associated macrophages promoted proliferation, migration, invasiveness, and breast tumor growth of ER+ cells, rendering these estrogen-dependent breast cancer cells resistant to estrogen withdrawal and tamoxifen or ICI 182,780 treatment. Crosstalk between breast cancer cells and conditioned macrophages induced sustained release of pro-inflammatory cytokines from both cell types, activation of NF-κB/STAT3/ERK in the cancer cells and hyperphosphorylation of ERα, which resulted constitutively active. Our simulated tumor microenvironment strongly altered endocrine and inflammatory signaling pathways in breast cancer cells, leading to endocrine resistance in these cells.
Collapse
|
43
|
Chen Y, Zheng K, Chen Z, Feng H, Fang W, Huang Z. [ADAM17 knockdown increases sensitivity of SW480 cells to cetuximad]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1366-1371. [PMID: 30514687 DOI: 10.12122/j.issn.1673-4254.2018.11.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To explore the association between expression of ADAM17 and cetuximad resistance in human colorectal cancer SW480 cells. METHODS The expression of ADAM17 was detected using Western blotting in different human colorectal cancer cell lines, and the cells highly expressing ADAM17 were selected as the target cells. SW480 cells were transfected with ADAM17-siRNA 1 and ADAM17-siRNA 2 and the changes in the expression of ADAM17 protein were detected using Western blotting. SW480 cells were exposed to cetuximad for 24 h and the cell apoptosis was analyzed using flow cytometry. Transwell assay was used to examine the migration ability of SW480 cells with different expression levels of ADAM17; Western blotting was used to analyze the changes in the expressions of AKT signaling pathway-related proteins in the treated cells. RESULTS The baseline expressions of ADAM17 were significantly higher in SW480 cells than in the other human colorectal cancer cell lines tested (P < 0.05). Both ADAM17-siRNA 1 and 2 effectively reduced the expression of ADAM17 protein in SW480 cells. Knockdown of ADAM17 with siRNA 1 significantly increased the sensitivity of SW480 cells to tocetuximad (P < 0.05), obviously inhibited the cell proliferation, migration and invasion, and significantly reduced the expressions of p-EGFR and p-AKT in the cells (P < 0.001). CONCLUSIONS ADAM17 knockdown obviously inhibits EGFR-AKT signaling pathway and increases the sensitivity of SW480 cells to tocetuximad.
Collapse
Affiliation(s)
- Ying Chen
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Kehong Zheng
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zetao Chen
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haizhan Feng
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Wei Fang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zonghai Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
44
|
Radhakrishnan R, Ha JH, Jayaraman M, Liu J, Moxley KM, Isidoro C, Sood AK, Song YS, Dhanasekaran DN. Ovarian cancer cell-derived lysophosphatidic acid induces glycolytic shift and cancer-associated fibroblast-phenotype in normal and peritumoral fibroblasts. Cancer Lett 2018; 442:464-474. [PMID: 30503552 DOI: 10.1016/j.canlet.2018.11.023] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/16/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022]
Abstract
Cancer-associated fibroblasts (CAFs) play a critical role in cancer progression, metastasis, and therapy resistance. Molecular events that confer CAF-phenotype to predecessor-cells are not fully understood. We demonstrate here that the ovarian cancer cell-conditioned medium (OCC-CM) induces CAF-phenotype in MRC5 lung-fibroblasts and it can be mimicked by LPA. While OCC-CM and LPA stimulated the expression of cellular CAF-markers by 3-days, they induced aerobic glycolysis, a metabolic marker for CAF, by 6 hrs. OCC-CM/LPA-induced glycolysis in lung (MRC5) as well as ovarian fibroblasts (NOF151) was inhibited by the LPA-receptor antagonist, Ki16425. Ovarian cancer patient-derived ascitic fluid-induced aerobic glycolysis in both NFs and Ovarian CAFs and it was inhibited by Ki16425. Further analysis indicated that LPA upregulated HIF1α-levels and the silencing of HIF1α attenuated LPA-induced glycolysis in both NOFs and CAFs. These results establish LPA-induced glycolytic-shift as the earliest, potentially priming event, in NF to CAF-transition. These findings also identify a role for LPA-LPAR-HIF1α signaling-hub in the maintenance of the glycolytic-phenotype in CAFs. Our results provide evidence that targeted inhibition of LPA-mediated metabolic reprogramming in CAFs may represent an adjuvant therapy in ovarian cancer.
Collapse
Affiliation(s)
| | - Ji Hee Ha
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, USA
| | - Muralidharan Jayaraman
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Katherine M Moxley
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics & Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Ciro Isidoro
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Anil K Sood
- Department of Gynecologic Oncology & Reproductive Medicine, and the Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yong Sang Song
- Cancer Research Institute, Seoul National University, College of Medicine, Seoul, 151-921, South Korea
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, USA.
| |
Collapse
|
45
|
Role of mTOR Signaling in Tumor Microenvironment: An Overview. Int J Mol Sci 2018; 19:ijms19082453. [PMID: 30126252 PMCID: PMC6121402 DOI: 10.3390/ijms19082453] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/06/2018] [Accepted: 08/15/2018] [Indexed: 12/31/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) pathway regulates major processes by integrating a variety of exogenous cues, including diverse environmental inputs in the tumor microenvironment (TME). In recent years, it has been well recognized that cancer cells co-exist and co-evolve with their TME, which is often involved in drug resistance. The mTOR pathway modulates the interactions between the stroma and the tumor, thereby affecting both the tumor immunity and angiogenesis. The activation of mTOR signaling is associated with these pro-oncogenic cellular processes, making mTOR a promising target for new combination therapies. This review highlights the role of mTOR signaling in the characterization and the activity of the TME’s elements and their implications in cancer immunotherapy.
Collapse
|
46
|
Abstract
Cancer is a major cause of death. The diversity of cancer types and the propensity of cancers to acquire resistance to therapies, including new molecularly targeted and immune-based therapies, drives the search for new ways to understand cancer progression. The remodelling of calcium (Ca2+) signalling and the role of the Ca2+ signal in controlling key events in cancer cells such as proliferation, invasion and the acquisition of resistance to cell death pathways is well established. Most of the work defining such changes has focused on Ca2+ permeable Transient Receptor Potential (TRP) Channels and some voltage gated Ca2+ channels. However, the identification of ORAI channels, a little more than a decade ago, has added a new dimension to how a Ca2+ influx pathway can be remodelled in some cancers and also how calcium signalling could contribute to tumour progression. ORAI Ca2+ channels are now an exemplar for how changes in the expression of specific isoforms of a Ca2+ channel component can occur in cancer, and how such changes can vary between cancer types (e.g. breast cancer versus prostate cancer), and even subtypes (e.g. oestrogen receptor positive versus oestrogen receptor negative breast cancers). ORAI channels and store operated Ca2+ entry are also highlighting the diverse roles of Ca2+ influx pathways in events such as the growth and metastasis of cancers, the development of therapeutic resistance and the contribution of tumour microenvironmental factors in cancer progression. In this review we will highlight some of the studies that have provided evidence for the need to deepen our understanding of ORAI Ca2+ channels in cancer. Many of these studies have also suggested new ways on how we can exploit the role of ORAI channels in cancer relevant processes to develop or inform new therapeutic strategies.
Collapse
|
47
|
Fiore APZP, Ribeiro PDF, Bruni-Cardoso A. Sleeping Beauty and the Microenvironment Enchantment: Microenvironmental Regulation of the Proliferation-Quiescence Decision in Normal Tissues and in Cancer Development. Front Cell Dev Biol 2018; 6:59. [PMID: 29930939 PMCID: PMC6001001 DOI: 10.3389/fcell.2018.00059] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/18/2018] [Indexed: 01/18/2023] Open
Abstract
Cells from prokaryota to the more complex metazoans cease proliferating at some point in their lives and enter a reversible, proliferative-dormant state termed quiescence. The appearance of quiescence in the course of evolution was essential to the acquisition of multicellular specialization and compartmentalization and is also a central aspect of tissue function and homeostasis. But what makes a cell cease proliferating even in the presence of nutrients, growth factors, and mitogens? And what makes some cells "wake up" when they should not, as is the case in cancer? Here, we summarize and discuss evidence showing how microenvironmental cues such as those originating from metabolism, extracellular matrix (ECM) composition and arrangement, neighboring cells and tissue architecture control the cellular proliferation-quiescence decision, and how this complex regulation is corrupted in cancer.
Collapse
Affiliation(s)
| | | | - Alexandre Bruni-Cardoso
- e-Signal Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
48
|
Recapitulating spatiotemporal tumor heterogeneity in vitro through engineered breast cancer microtissues. Acta Biomater 2018; 73:236-249. [PMID: 29679778 DOI: 10.1016/j.actbio.2018.04.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 01/14/2023]
Abstract
Tumor and microenvironmental heterogeneity hinders the study of breast cancer biology and the assessment of therapeutic strategies, being associated with high variability and drug resistance. In this context, it is mandatory to develop three-dimensional breast tumor models able to reproduce this heterogeneity and the dynamic interaction occurring between tumor cells and microenvironment. Here we show a new breast cancer microtissue model (T-µTP) uniquely able to present intra-tumor morphological heterogeneity in a dynamic and responsive endogenous matrix. T-µTP consists of adenocarcinoma cells, endothelial cells and stromal fibroblasts. These three kinds of cells are totally embedded into an endogenous matrix which is rich in collagen and hyaluronic acid and it is directly produced by human fibroblasts. In this highly physiologically relevant environment, tumor cells evolve in different cluster morphologies recapitulating tumor spatiotemporal heterogeneity. Moreover they activate the desmoplastic and vascular reaction with affected collagen content, assembly and organization and the presence of aberrant capillary-like structures (CLS). Thus, T-µTP allows to outline main crucial events involved in breast cancer progression into a single model overcoming the limit of artificial extra cellular matrix surrogates. We strongly believe that T-µTP is a suitable model for the study of breast cancer and for drug screening assays following key parameters of clinical interest. STATEMENT OF SIGNIFICANCE Tumor and microenvironmental heterogeneity makes very hurdle to find a way to study and treat breast cancer. Here we develop an innovative 3D tumor microtissue model recapitulating in vitro tumor heterogeneity. Tumor microtissues are characterized by the activation of the stromal and vascular reaction too. We underline the importance to mimic different microenvironmental tumor features in the same time and in a single tissue in order to obtain a model of spatiotemporal tumor genesis and progression, suitable for the study of tumor treatment and resistance.
Collapse
|
49
|
Calcium signaling and the therapeutic targeting of cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1786-1794. [PMID: 29842892 DOI: 10.1016/j.bbamcr.2018.05.015] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/23/2018] [Accepted: 05/24/2018] [Indexed: 12/14/2022]
Abstract
The calcium signal is implicated in a variety of processes important in tumor progression (e.g. proliferation and invasiveness). The calcium signal has also been shown to be important in other processes important in cancer progression including the development of resistance to current cancer therapies. In this review, we discuss how Ca2+ channels, pumps and exchangers may be drug targets in some cancer types. We consider what factors should be taken into account when considering an optimal Ca2+ channel, pump or exchanger as a candidate for further assessment as a novel drug target in cancer. We also present and summarize how some therapies for the treatment of cancer intersect with Ca2+ signaling and how pharmacological manipulation of the machinery of Ca2+ signaling could promote the effectiveness of some therapies. We also review new therapeutic opportunities for Ca2+ signal modulators in the context of the tumor microenvironment.
Collapse
|
50
|
Bergamo A, Dyson PJ, Sava G. The mechanism of tumour cell death by metal-based anticancer drugs is not only a matter of DNA interactions. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.01.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|