1
|
Robles-Remacho A, Nilsson M. Spatial miRNomics: towards the integration of microRNAs in spatial biology. Nat Rev Genet 2025; 26:291-292. [PMID: 39915688 DOI: 10.1038/s41576-025-00819-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Affiliation(s)
- Agustín Robles-Remacho
- Science For Life Laboratory (SciLifeLab), Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden.
| | - Mats Nilsson
- Science For Life Laboratory (SciLifeLab), Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden.
| |
Collapse
|
2
|
Chen L, Zhu M, Zhang H, Tang W, Liu J, Tang X, Chang X, Zhu Z, Liu T, Weng L, Zhang Y, Chen X. Remodeling of Effector and Regulatory T Cells by Capture and Utilization of miRNAs Using Nanocomposite Hydrogel for Tumor-Specific Photothermal Immunotherapy. ACS NANO 2025; 19:14873-14892. [PMID: 40223522 DOI: 10.1021/acsnano.4c18801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
In immunotherapy for malignant tumors, the dysregulation of the balance between effector T cells and regulatory T cells (Tregs) and the uncertain efficacy due to individual differences have been considered as two critical challenges. In this study, we engineered an injectable nanocomposite hydrogel system (SNAs@M-Gel) capable of suppressing Treg proliferation and blocking PD-1/PD-L1-mediated immune evasion effectively, achieved through the stimulus-responsive modulation of multiple tumor-associated microRNAs. Simultaneously, this system enables microRNA-dependent photothermal immunotherapy, facilitating a highly efficient and personalized approach to tumor treatment. Specifically, oxidized sodium alginate (OSA) and cancer cell membrane (CCM)-encapsulated spherical nucleic acid nanoparticles (SNAs@M) were used to construct the SNAs@M-Gel hydrogel in situ at the tumor site through the formation of pH-sensitive Schiff base bonding and cross-linking using endogenous calcium ions (Ca2+). During treatment, SNAs@M-Gel was retained locally for up to 10 days, and SNAs@M nanoparticles were continuously released into the tumor microenvironment. Through the targeting ability of CCM, SNAs@M precisely entered tumor cells and specifically hybridized with the overexpressed miR-214 and miR-130a, leading to a significant downregulation of PD-L1 expression on tumor cells and the restoration of cytotoxic T lymphocyte (CTL) function suppressed by Tregs, thereby remodeling the immune microenvironment. In addition, miRNAs functioned as cross-linking agents, facilitating the aggregation of SNAs and allowing the localized production of photothermal agents directly inside tumor cells, which, under near-infrared (NIR) irradiation, promoted highly selective photothermal therapy. This cascade of events not only led to the destruction of the primary tumor but also resulted in the release of a substantial number of tumor-related antigens, which triggered the maturation of adjacent dendritic cells (DCs) and subsequent priming of tumor-specific CTLs, while simultaneously depleting Tregs, thereby reversing the tumor-promoting immune microenvironment and enhancing the overall therapeutic efficacy of photothermal immunotherapy.
Collapse
Affiliation(s)
- Li Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Handan Zhang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Wenjun Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Jie Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Xiaoyu Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Xiaowei Chang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Tao Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Lin Weng
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| |
Collapse
|
3
|
Chen L, Tang W, Liu J, Zhu M, Mu W, Tang X, Liu T, Zhu Z, Weng L, Cheng Y, Zhang Y, Chen X. On-demand reprogramming of immunosuppressive microenvironment in tumor tissue via multi-regulation of carcinogenic microRNAs and RNAs dependent photothermal-immunotherapy using engineered gold nanoparticles for malignant tumor treatment. Biomaterials 2025; 315:122956. [PMID: 39549441 DOI: 10.1016/j.biomaterials.2024.122956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
The frequent immune escape of tumor cells and fluctuating therapeutic efficiency vary with each individual are two critical issues for immunotherapy against malignant tumor. Herein, we fabricated an intelligent core-shell nanoparticle (SNAs@CCMR) to significantly inhibit the PD-1/PD-L1 mediated immune escape by on-demand regulation of various oncogenic microRNAs and perform RNAs dependent photothermal-immunotherapy to achieve precise and efficient treatment meeting the individual requirements of specific patients by in situ generation of customized tumor-associated antigens. The SNAs@CCMR consisted of antisense oligonucleotides grafted gold nanoparticles (SNAs) as core and TLR7 agonist imiquimod (R837) functionalized cancer cell membrane (CCM) as shell, in which the acid-labile Schiff base bond was used to connect the R837 and CCM. During therapy, the acid environment of tumor tissue cleaved the Schiff base to generate free R837 and SNAs@CCM. The SNAs@CCM further entered tumor cells via CCM mediated internalization, and then specifically hybridized with over-expressed miR-130a and miR-21, resulting in effective inhibition of the migration and PD-L1 expression of tumor cells to avoid their immune escape. Meanwhile, the RNAs capture also caused significant aggregation of SNAs, which immediately generated photothermal agents within tumor cells to perform highly selective photothermal therapy under NIR irradiation. These chain processes not only damaged the primary tumor, but also produced plenty of tumor-associated antigens, which matured the surrounding dendritic cells (DCs) and activated anti-tumor T cells along with the released R837, resulting in the enhanced immunotherapy with suppressive immune escape. Both in vivo and in vitro experiments demonstrated that our nanoparticles were able to inhibit primary tumor and its metastasis via multi-regulation of carcinogenic microRNAs and RNAs dependent photothermal-immune activations, which provided a promising strategy to reprogram the immunosuppressive microenvironment in tumor tissue for better malignant tumor therapy.
Collapse
Affiliation(s)
- Li Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Wenjun Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jie Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wenyun Mu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiaoyu Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tao Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lin Weng
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yumeng Cheng
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
4
|
Makada H, Singh M. Hydrogels as Suitable miRNA Delivery Systems: A Review. Polymers (Basel) 2025; 17:915. [PMID: 40219305 PMCID: PMC11991254 DOI: 10.3390/polym17070915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
The use of miRNA in therapeutics has, since its discovery in 1993, attracted tremendous attention, and research in this area has progressed rapidly. Since the advent of RNA interference (RNAi), much about the nucleic acid siRNA has been elucidated. At the same time, no miRNA-based drugs have passed phase II clinical trials. A significant obstacle to miRNA-based drug development is the ease of degradation and relatively short half-life in vivo of miRNA. Hydrogels are networks of cross-linked polymer chains with the ability to 'swell'. They have shown remarkable capabilities that improve the properties of other researched carriers. In combination with miRNA modification strategies and inorganic carriers, hydrogel systems show promise for sustained miRNA delivery and the development of novel miRNA-based drugs. Although hydrogel systems have been reported recently, the focus has been predominantly on their wound-healing properties, with a dearth of information on their nucleic acid carrier abilities. This paper focuses more on the latest advancements in developing hydrogels as a carrier system, emphasizing the delivery of miRNA. This review will cover the methods of hydrogel fabrication, efforts for sustained miRNA release, biomedical applications, and future prospects.
Collapse
Affiliation(s)
| | - Moganavelli Singh
- Nano-Gene and Drug Delivery Laboratory, Discipline of Biochemistry, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa;
| |
Collapse
|
5
|
Strati A, Adamopoulos C, Kotsantis I, Psyrri A, Lianidou E, Papavassiliou AG. Targeting the PD-1/PD-L1 Signaling Pathway for Cancer Therapy: Focus on Biomarkers. Int J Mol Sci 2025; 26:1235. [PMID: 39941003 PMCID: PMC11818137 DOI: 10.3390/ijms26031235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
The PD1/PD-L1 axis plays an important immunosuppressive role during the T-cell-mediated immune response, which is essential for the physiological homeostasis of the immune system. The biology of the immunological microenvironment is extremely complex and crucial for the development of treatment strategies for immunotherapy. Characterization of the immunological, genomic or transcriptomic landscape of cancer patients could allow discrimination between responders and non-responders to anti-PD-1/PD-L1 therapy. Immune checkpoint inhibitor (ICI) therapy has shown remarkable efficacy in a variety of malignancies in landmark trials and has fundamentally changed cancer therapy. Current research focuses on strategies to maximize patient selection for therapy, clarify mechanisms of resistance, improve existing biomarkers, including PD-L1 expression and tumor mutational burden (TMB), and discover new biomarkers. In this review, we focus on the function of the PD-1/PD-L1 signaling pathway and discuss the immunological, genomic, epigenetic and transcriptomic landscape in cancer patients receiving anti-PD-1/PD-L1 therapy. Finally, we provide an overview of the clinical trials testing the efficacy of antibodies against PD-1/PD-L1.
Collapse
Affiliation(s)
- Areti Strati
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece;
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (A.G.P.)
| | - Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (A.G.P.)
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ioannis Kotsantis
- Department of Medical Oncology, Second Department of Internal Medicine, Attikon University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Amanda Psyrri
- Department of Medical Oncology, Second Department of Internal Medicine, Attikon University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (A.G.P.)
| |
Collapse
|
6
|
Peng HY, Chang CW, Wu PH, Li LJ, Lin YL, Hsiao M, Chang JY, Chang PMH, Lee HL, Chang WM. Oral Cancer-Derived miR-762 Suppresses T-Cell Infiltration and Activation by Horizontal Inhibition of CXCR3 Expression. Int J Mol Sci 2025; 26:1077. [PMID: 39940842 PMCID: PMC11817288 DOI: 10.3390/ijms26031077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is an immune-cold tumor characterized by an immunosuppressive microenvironment with low cytotoxic activity to eliminate tumor cells. Tumor escape is one of the initial steps in cancer development. Understanding the underlying mechanisms of cancer escape can help researchers develop new treatment strategies. In this study, we prove the oral oncogenic miR-762 can suppress T-cell recruitment and cytotoxic activation in the tumor microenvironment (TME) through horizontal transmission from OSCC cells to adaptive immune T cells. Public database analysis and quantitative real-time polymerase chain reaction (qRT-PCR) were used to determine the prognosis and expression of miR-762 in OSCC. T-cell activation by flow cytometry, qRT-PCR, IL-12 secretion, and T-cell recruitment and cytotoxicity abilities were conducted in the miR-762 manipulation T-cell and OSCC-T-cell co-culture system. A luciferase reporter and CXCR3 protein expression were also carried out to validate the direct interaction between CXCR3 and microRNA (miR)-762. This horizontal transmission of miR-762 directly suppresses CXCR3 expression in T cells, inhibiting CXCR3-induced T-cell migration and downstream T-cell cytotoxic activity by disrupting AKT activation. Additionally, miR-762 transmission suppressed T-cell activation marker expression, T-cell proliferation, IL-12 secretion, and T-cell cytotoxicity. In conclusion, our findings reveal a novel miR-762/CXCR3 axis that regulates the immunosuppressive microenvironment in OSCC and may be a potential RNA-targeted therapeutic approach to restore the anti-tumor immune response in OSCC treatment.
Collapse
Affiliation(s)
- Hsuan-Yu Peng
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Research Center of Oral Translational Medicine, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Chia-Wei Chang
- Division of Family Dentistry, Department of Dentistry, Taipei Medical University Hospital, Taipei 110, Taiwan;
| | - Ping-Hsiu Wu
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan
- TMU Proton Center, Taipei Medical University, Taipei 110, Taiwan
| | - Li-Jie Li
- Ph.D. Program of School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Oral Pathology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Yu-Lung Lin
- The Ph.D. Program for Translational Medicine, College of Medical Sciences and Technology, Taipei Medical University, Taipei 110, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Jang-Yang Chang
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Peter Mu-Hsin Chang
- Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan;
- Faculty of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hsin-Lun Lee
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan
- TMU Proton Center, Taipei Medical University, Taipei 110, Taiwan
| | - Wei-Min Chang
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Research Center of Oral Translational Medicine, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
7
|
Mohseni A, Salehi F, Rostami S, Hadiloo K, Hashemi M, Baridjavadi Z, Ahangari F, Karami N, Samani F, Tahmasebi S, Farahani N, Taheriazam A. Harnessing the power of exosomes for diagnosis, prognosis, and treatment of hematological malignancies. Stem Cell Res Ther 2025; 16:6. [PMID: 39773361 PMCID: PMC11708188 DOI: 10.1186/s13287-024-04125-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
Exosomes are small extracellular vesicles of endocytic origin released by various cell types. They consist of lipid bilayers containing macromolecules such as lipids, proteins, microRNAs, growth factors, cytokines, and carbohydrates. Exosomes play a critical role in the diagnosis and treatment of various diseases. For instance, exosome contents have been utilized as biomarkers in body fluids (urine, saliva, serum) to identify cancers, autoimmune diseases, and inflammatory conditions such as sepsis. Due to their small size and ability to reach tumor microenvironments, exosomes are also used as carriers for chemotherapeutic drugs in drug delivery systems. Furthermore, evidence indicates that malignant cells release exosomes into the tumor microenvironment, influencing immune cells in a paracrine manner. Additionally, immune cell-derived exosomes, such as those from Natural Killer (NK) cells or cytotoxic T lymphocytes (CTLs), show potential as therapeutic agents in treating malignancies like leukemia. This review discusses the diagnostic role of exosomes in various hematological malignancies and explores the therapeutic potential of immune cell-derived exosomes in these diseases.
Collapse
Affiliation(s)
- Amirata Mohseni
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Fatemeh Salehi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Samaneh Rostami
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kaveh Hadiloo
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Baridjavadi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Ahangari
- Department of Immunology, Pasteur Institue of Iran, Tehran, Iran
| | - Najibeh Karami
- Hematology-Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Samani
- Blood Transfusion Research Center, High Institute for Research and Education in transfusion medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
8
|
Lefler DS, Manobianco SA, Bashir B. Immunotherapy resistance in solid tumors: mechanisms and potential solutions. Cancer Biol Ther 2024; 25:2315655. [PMID: 38389121 PMCID: PMC10896138 DOI: 10.1080/15384047.2024.2315655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
While the emergence of immunotherapies has fundamentally altered the management of solid tumors, cancers exploit many complex biological mechanisms that result in resistance to these agents. These encompass a broad range of cellular activities - from modification of traditional paradigms of immunity via antigen presentation and immunoregulation to metabolic modifications and manipulation of the tumor microenvironment. Intervening on these intricate processes may provide clinical benefit in patients with solid tumors by overcoming resistance to immunotherapies, which is why it has become an area of tremendous research interest with practice-changing implications. This review details the major ways cancers avoid both natural immunity and immunotherapies through primary (innate) and secondary (acquired) mechanisms of resistance, and it considers available and emerging therapeutic approaches to overcoming immunotherapy resistance.
Collapse
Affiliation(s)
- Daniel S. Lefler
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven A. Manobianco
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Babar Bashir
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
9
|
Wu Y, Xiao Y, Ding Y, Ran R, Wei K, Tao S, Mao H, Wang J, Pang S, Shi J, Zhu C, Wan W, Yang Q, Chen C. Colorectal cancer cell-derived exosomal miRNA-372-5p induces immune escape from colorectal cancer via PTEN/AKT/NF-κB/PD-L1 pathway. Int Immunopharmacol 2024; 143:113261. [PMID: 39353381 DOI: 10.1016/j.intimp.2024.113261] [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: 06/21/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Tumor cells can escape immune surveillance by changing their own escape or expressing abnormal genes and proteins, resulting in unlimited proliferation and invasive growth of cells. These changes are related to microRNAs (miRNAs), which reduce the killing effect of immune cells, devastate the immune response, and interfere with apoptosis through the aberrant expression of relevant miRNAs. In the preliminary phase of this study, miRNAs in clinical plasma exosomes of colorectal cancer patients were differentially analyzed by RNA sequencing technology, and miR-372-5p derived from extracellular vesicles (sEVs) was found to be a key signaling molecule mediating the regulation of macrophages by colorectal cancer (CRC). miRNA-372-5p is upregulated in colorectal cancer patient tissues and serum, as well as colorectal cancer cell lines and their exosomes. Subsequently, we found that macrophages could take up sEV secreted by colorectal cancer cells HCT116, affecting the expression of the immune checkpoint PD-L1, resulting in the generation of a tumor-immunosuppressive microenvironment and suppression of T cell activation in CRC. Gene enrichment mapping and database revealed that miR-372-5p regulates PD-L1 expression in colorectal cancer through the homologous phosphatase-tensin (PTEN)-phosphatidylinositol 3-kinase-protein kinase B (AKT)-nuclear factor-κB (NF-κB) pathway. Further studies confirmed that miRNA-372-5p-treated macrophages co-cultured with T cells affected the regulation of PD-L1 expression through the PTEN/AKT/NF-κB signaling pathway, resulting in decreased CD3+CD8+ T cell activity, decreased cytokine IL-2 and increased IFN-γ. And miRNA-372-5p could down-regulate the expression of PD-L1 in HCT116 through the PTEN/AKT/NF-κB pathway, inhibit tumor cell proliferation and promote apoptosis. Conclusion: Colorectal cancer cell-derived exosome miR-372-5p can be phagocytosed by colorectal cancer and macrophage cells, regulate the expression of PD-L1 in colorectal cancer cells and macrophages by targeting the PTEN/AKT/NF-κB pathway, and induce the immunosuppressive microenvironment of CRC to promote CRC development. This suggests that inhibiting the secretion of HCT116-specific sEV-miR-372-5p or targeting PD-L1 in tumor-associated macrophages could be a novel approach for CRC treatment and possibly a sensitizing approach for CRC anti-PD-L1 therapy.
Collapse
Affiliation(s)
- Yulun Wu
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China; Department of Life Sciences, Bengbu Medical University, Anhui 233030, China.
| | - Yuhan Xiao
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China; School of Laboratory Medicine, Bengbu Medical University, Anhui 233030, China.
| | - Yongxing Ding
- The Third the Pople's Hospital of Bengbu, Anhui 233000, China.
| | - Ruorong Ran
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China.
| | - Ke Wei
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China.
| | - Shuang Tao
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China.
| | - Huilan Mao
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China.
| | - Jing Wang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China.
| | - Siyan Pang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China.
| | - Jiwen Shi
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China.
| | - Chengle Zhu
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu medical university, 2600 Donghai Avenue, Bengbu, Anhui 233030, China.
| | - Wenrui Wan
- Department of Biotechnology, Bengbu Medical University, Anhui 233030, China.
| | - Qingling Yang
- Department of Biochemistry and Molecular Biology, Bengbu Medical University, Anhui 233030, China.
| | - Changjie Chen
- Department of Biochemistry and Molecular Biology, Bengbu Medical University, Anhui 233030, China.
| |
Collapse
|
10
|
Karabay AZ, Ozkan T, Karadag Gurel A, Koc A, Hekmatshoar Y, Sunguroglu A, Aktan F, Buyukbingöl Z. Identification of exosomal microRNAs and related hub genes associated with imatinib resistance in chronic myeloid leukemia. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9701-9721. [PMID: 38916832 PMCID: PMC11582232 DOI: 10.1007/s00210-024-03198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024]
Abstract
Chemotherapy resistance is a major obstacle in cancer therapy, and identifying novel druggable targets to reverse this phenomenon is essential. The exosome-mediated transmittance of drug resistance has been shown in various cancer models including ovarian and prostate cancer models. In this study, we aimed to investigate the role of exosomal miRNA transfer in chronic myeloid leukemia drug resistance. For this purpose, firstly exosomes were isolated from imatinib sensitive (K562S) and resistant (K562R) chronic myeloid leukemia (CML) cells and named as Sexo and Rexo, respectively. Then, miRNA microarray was used to compare miRNA profiles of K562S, K562R, Sexo, Rexo, and Rexo-treated K562S cells. According to our results, miR-125b-5p and miR-99a-5p exhibited increased expression in resistant cells, their exosomes, and Rexo-treated sensitive cells compared to their sensitive counterparts. On the other hand, miR-210-3p and miR-193b-3p were determined to be the two miRNAs which exhibited decreased expression profile in resistant cells and their exosomes compared to their sensitive counterparts. Gene targets, signaling pathways, and enrichment analysis were performed for these miRNAs by TargetScan, KEGG, and DAVID. Potential interactions between gene candidates at the protein level were analyzed via STRING and Cytoscape software. Our findings revealed CCR5, GRK2, EDN1, ARRB1, P2RY2, LAMC2, PAK3, PAK4, and GIT2 as novel gene targets that may play roles in exosomal imatinib resistance transfer as well as mTOR, STAT3, MCL1, LAMC1, and KRAS which are already linked to imatinib resistance. MDR1 mRNA exhibited higher expression in Rexo compared to Sexo as well as in K562S cells treated with Rexo compared to K562S cells which may suggest exosomal transfer of MDR1 mRNA.
Collapse
Affiliation(s)
- Arzu Zeynep Karabay
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey.
| | - Tulin Ozkan
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey.
| | - Aynur Karadag Gurel
- Department of Medical Biology, Faculty of Medicine, Usak University, Usak, Turkey.
| | - Asli Koc
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Yalda Hekmatshoar
- Department of Medical Biology, Faculty of Medicine, Altinbas University, Istanbul, Turkey
| | - Asuman Sunguroglu
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Fugen Aktan
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Zeliha Buyukbingöl
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
11
|
Lv Y, Gai K, Ding X, Sun W. Soluble forms of PD-1 and sPD-L1/2 in serum and urine of patients with head and neck cancer and their clinical significance. Biotechnol Genet Eng Rev 2024; 40:2234-2245. [PMID: 37057626 DOI: 10.1080/02648725.2023.2199237] [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: 02/20/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
This study explored the soluble forms of PD-1 and sPD-L1/2 in serum and urine of patients with head and neck cancer (HNCs) and associated the data with clinical state and 5-year survival. The sPD-1 and sPD-L1/2 levels were evaluated by ELISA in sufferers (N=110) and normal controls (N=82). Patients in the case group were more likely to be male smokers or former smokers. Compared with the normal control group, the serum levels of sPD-1, sPD-L1 and sPD-L2 and the urine level of sPD-L1 in patients with HNCs were increased. Furthermore, sPD-1 and sPD-L1 serum levels existed a positive connection, and sPD-1 and sPD-L2 serum levels positively correlated in HNCs sufferers. The urine sPD-1 and sPD-L1 had a positive relationship. sPD-1 serum levels had a positive connection with urine sPD-1, sPD-L1 urine levels had a positive relationship with sPD-L1, and sPD-L2 serum levels positively connected to urine sPD-L2. Lower serum sPD-1 and sPD-L1/L2 were associated with disease progression and survival at the examination time. sPD-1 and sPD-L1/L2 serum levels above median were markedly related to a decreased probability of 5-years OS in patients with HNCs. The sPD-1 and sPD-L1/2 were complementary markers representing clinical condition and illness outcomes for HNCs patients. The sPD-L1 might accelerate the characterization of high-risk patients with disapproving illness outcomes. sPD-1 and sPD-L1/2 could be easily accessed through liquid biopsy. The incorporation of them as indicators for risk evaluation throughout treatment scheduling and follow-up seems to be an appreciated method.
Collapse
Affiliation(s)
- Yan Lv
- Department of Eastern Hospital Oncology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Kai Gai
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
| | - Xia Ding
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
| | - Weihua Sun
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
| |
Collapse
|
12
|
Sharma A, Vikramdeo KS, Sudan SK, Anand S, Deshmukh SK, Singh AP, Singh S. Cortisol affects macrophage polarization by inducing miR-143/145 cluster to reprogram glucose metabolism and by promoting TCA cycle anaplerosis. J Biol Chem 2024; 300:107753. [PMID: 39260692 PMCID: PMC11470657 DOI: 10.1016/j.jbc.2024.107753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
Chronic stress can have adverse consequences on human health by disrupting the hormonal balance in our body. Earlier, we observed elevated levels of cortisol, a primary stress hormone, and some exosomal microRNAs in the serum of patients with breast cancer. Here, we investigated the role of cortisol in microRNA induction and its functional consequences. We found that cortisol induced the expression of miR-143/145 cluster in human monocyte (THP1 and U937)-derived macrophages but not in breast cancer cells. In silico analysis identified glucocorticoid-response element in the upstream CARMN promoter utilized by the miR-143/145 cluster. Enhanced binding of glucocorticoid-receptor (GR) upon cortisol exposure and its regulatory significance was confirmed by chromatin-immunoprecipitation and promoter-reporter assays. Further, cortisol inhibited IFNγ-induced M1 polarization and promoted M2 polarization, and these effects were suppressed by miR-143-3p and miR-145-5p inhibitors pretreatment. Cortisol-treated macrophages exhibited increased oxygen-consumption rate (OCR) to extracellular-acidification rate (ECAR) ratio, and this change was neutralized by functional inhibition of miR-143-3p and miR-145-5p. HK2 and ADPGK were confirmed as the direct targets of miR-143-3p and miR-145-5p, respectively. Interestingly, silencing of HK2 and ADPGK inhibited IFNγ-induced M1 polarization but failed to induce M2 polarization, since it suppressed both ECAR and OCR, while OCR was largely sustained in cortisol-treated M2-polarized macrophages. We found that cortisol treatment sustained OCR by enhancing fatty acid and glutamine metabolism through upregulation of CPT2 and GLS, respectively, to support M2 polarization. Thus, our findings unfold a novel mechanism of immune suppression by cortisol and open avenues for preventive and therapeutic interventions.
Collapse
Affiliation(s)
- Amod Sharma
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi, USA; Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kunwar Somesh Vikramdeo
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi, USA; Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Sarabjeet Kour Sudan
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi, USA; Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Shashi Anand
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi, USA; Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Sachin Kumar Deshmukh
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA; Department of Pathology, University of South Alabama, Mobile, Alabama, USA
| | - Ajay Pratap Singh
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi, USA; Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Seema Singh
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi, USA; Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA.
| |
Collapse
|
13
|
Yadav R, Khatkar R, Yap KCH, Kang CYH, Lyu J, Singh RK, Mandal S, Mohanta A, Lam HY, Okina E, Kumar RR, Uttam V, Sharma U, Jain M, Prakash H, Tuli HS, Kumar AP, Jain A. The miRNA and PD-1/PD-L1 signaling axis: an arsenal of immunotherapeutic targets against lung cancer. Cell Death Discov 2024; 10:414. [PMID: 39343796 PMCID: PMC11439964 DOI: 10.1038/s41420-024-02182-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Lung cancer is a severe challenge to the health care system with intrinsic resistance to first and second-line chemo/radiotherapies. In view of the sterile environment of lung cancer, several immunotherapeutic drugs including nivolumab, pembrolizumab, atezolizumab, and durvalumab are currently being used in clinics globally with the intention of releasing exhausted T-cells back against refractory tumor cells. Immunotherapies have a limited response rate and may cause immune-related adverse events (irAEs) in some patients. Hence, a deeper understanding of regulating immune checkpoint interactions could significantly enhance lung cancer treatments. In this review, we explore the role of miRNAs in modulating immunogenic responses against tumors. We discuss various aspects of how manipulating these checkpoints can bias the immune system's response against lung cancer. Specifically, we examine how altering the miRNA profile can impact the activity of various immune checkpoint inhibitors, focusing on the PD-1/PD-L1 pathway within the complex landscape of lung cancer. We believe that a clear understanding of the host's miRNA profile can influence the efficacy of checkpoint inhibitors and significantly contribute to existing immunotherapies for lung cancer patients. Additionally, we discuss ongoing clinical trials involving immunotherapeutic drugs, both as standalone treatments and in combination with other therapies, intending to advance the development of immunotherapy for lung cancer.
Collapse
Affiliation(s)
- Ritu Yadav
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Rinku Khatkar
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Kenneth C-H Yap
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chloe Yun-Hui Kang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juncheng Lyu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rahul Kumar Singh
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Surojit Mandal
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Adrija Mohanta
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rajiv Ranjan Kumar
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Vivek Uttam
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Uttam Sharma
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Manju Jain
- Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, India
| | | | | | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Aklank Jain
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
14
|
Wang C, Zhang Y, Kong W, Rong X, Zhong Z, Jiang L, Chen S, Li C, Zhang F, Jiang J. Delivery of miRNAs Using Nanoparticles for the Treatment of Osteosarcoma. Int J Nanomedicine 2024; 19:8641-8660. [PMID: 39188861 PMCID: PMC11346496 DOI: 10.2147/ijn.s471900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
Osteosarcoma is the predominant primary malignant bone tumor that poses a significant global health challenge. MicroRNAs (miRNAs) that regulate gene expression are associated with osteosarcoma pathogenesis. Thus, miRNAs are potential therapeutic targets for osteosarcoma. Nanoparticles, widely used for targeted drug delivery, facilitate miRNA-based osteosarcoma treatment. Numerous studies have focused on miRNA delivery using nanoparticles to inhibit the progress of osteosarcoma. Polymer-based, lipid-based, inorganic-based nanoparticles and extracellular vesicles were used to deliver miRNAs for the treatment of osteosarcoma. They can be modified to enhance drug loading and delivery capabilities. Also, miRNA delivery was combined with traditional therapies, for example chemotherapy, to treat osteosarcoma. Consequently, miRNA delivery offers promising therapeutic avenues for osteosarcoma, providing renewed hope for patients. This review emphasizes the studies utilizing nanoparticles for miRNA delivery in osteosarcoma treatment, then introduced and summarized the nanoparticles in detail. And it also discusses the prospects for clinical applications.
Collapse
Affiliation(s)
- Chengran Wang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Yihong Zhang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Weihui Kong
- Department of Stomatology, the First Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Xin’ao Rong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Ziming Zhong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Lei Jiang
- Department of Geriatric Medicine, Changchun Central Hospital, Changchun, Jilin Province, People’s Republic of China
| | - Shuhan Chen
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Chuang Li
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Fuqiang Zhang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Jinlan Jiang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| |
Collapse
|
15
|
Yu X, Zhang Y, Luo F, Zhou Q, Zhu L. The role of microRNAs in the gastric cancer tumor microenvironment. Mol Cancer 2024; 23:170. [PMID: 39164671 PMCID: PMC11334576 DOI: 10.1186/s12943-024-02084-x] [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: 06/25/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the deadliest malignant tumors with unknown pathogenesis. Due to its treatment resistance, high recurrence rate, and lack of reliable early detection techniques, a majority of patients have a poor prognosis. Therefore, identifying new tumor biomarkers and therapeutic targets is essential. This review aims to provide fresh insights into enhancing the prognosis of patients with GC by summarizing the processes through which microRNAs (miRNAs) regulate the tumor microenvironment (TME) and highlighting their critical role in the TME. MAIN TEXT A comprehensive literature review was conducted by focusing on the interactions among tumor cells, extracellular matrix, blood vessels, cancer-associated fibroblasts, and immune cells within the GC TME. The role of noncoding RNAs, known as miRNAs, in modulating the TME through various signaling pathways, cytokines, growth factors, and exosomes was specifically examined. Tumor formation, metastasis, and therapy in GC are significantly influenced by interactions within the TME. miRNAs regulate tumor progression by modulating these interactions through multiple signaling pathways, cytokines, growth factors, and exosomes. Dysregulation of miRNAs affects critical cellular processes such as cell proliferation, differentiation, angiogenesis, metastasis, and treatment resistance, contributing to the pathogenesis of GC. CONCLUSIONS miRNAs play a crucial role in the regulation of the GC TME, influencing tumor progression and patient prognosis. By understanding the mechanisms through which miRNAs control the TME, potential biomarkers and therapeutic targets can be identified to improve the prognosis of patients with GC.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Medical Oncology, West China Hospital, Sichuan University, Sichuan Province, Cancer Center, Chengdu, 610041, People's Republic of China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, People's Republic of China
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Sichuan Province, No. 10 Qinyun Nan Street, Chengdu, 610041, People's Republic of China
| | - Yin Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Pulmonary Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fengming Luo
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Pulmonary Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qinghua Zhou
- Department of Medical Oncology, West China Hospital, Sichuan University, Sichuan Province, Cancer Center, Chengdu, 610041, People's Republic of China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, People's Republic of China.
| | - Lingling Zhu
- Department of Medical Oncology, West China Hospital, Sichuan University, Sichuan Province, Cancer Center, Chengdu, 610041, People's Republic of China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
16
|
Wang S, Mouliere F, Pegtel DM, Chamuleau MED. Turning the tide in aggressive lymphoma: liquid biopsy for risk-adapted treatment strategies. Trends Mol Med 2024; 30:660-672. [PMID: 38692937 DOI: 10.1016/j.molmed.2024.04.005] [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: 02/12/2024] [Revised: 03/30/2024] [Accepted: 04/04/2024] [Indexed: 05/03/2024]
Abstract
Diffuse large B cell lymphoma (DLBCL) exhibits significant biological and clinical heterogeneity that presents challenges for risk stratification and disease surveillance. Existing tools for risk stratification, including the international prognostic index (IPI), tissue molecular analyses, and imaging, have limited accuracy in predicting outcomes. The therapeutic landscape for aggressive lymphoma is rapidly evolving, and there is a pressing need to identify patients at risk of refractory or relapsed (R/R) disease in the context of personalized therapy. Liquid biopsy, a minimally invasive method for cancer signal detection, has been explored to address these challenges. We review advances in liquid biopsy strategies focusing on circulating nucleic acids in DLBCL patients and highlight their clinical potential. We also provide recommendations for biomarker-guided trials to support risk-adapted treatment modalities.
Collapse
Affiliation(s)
- Steven Wang
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Florent Mouliere
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Cancer Research UK National Biomarker Centre, University of Manchester, Wilmslow Road, Manchester, UK
| | - D Michiel Pegtel
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Martine E D Chamuleau
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Aslan ES, Aydin E. Investigating the role of let-7a microRNA in cisplatin sensitivity of A549 lung cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3979-3984. [PMID: 37991541 DOI: 10.1007/s00210-023-02858-y] [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: 10/19/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023]
Abstract
Lung cancer (LC) is a major cause of death worldwide, and cisplatin is commonly used as a chemotherapeutic drug for the treatment of LC. However, high doses of cisplatin can reduce its efficacy, leading to the need for new methods to increase LC cell sensitivity to this drug molecule. To overcome this problem, it is important to discover new methods to increase the sensitivity of LC cells to cisplatin. In this study, we investigated the use of anti-let-7a, a microRNA, to enhance the cisplatin sensitivity in A549 LC cells by comparing its effects with the commonly used oncogenes akt1 and pik3ca. The A549 cell line was transfected with anti-let-7a, and its effects were analyzed using functional assays. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) assay was used for the measurement of cell viability, and gene expression levels of cell death-associated genes, were analyzed by using quantitative real-time PCR (qRT-PCR). Results showed that anti-let-7a downregulation decreased the viability of A549 cells significantly compared to the control group in the presence of cisplatin. Moreover, the single treatment of cells with anti-let-7a and cisplatin resulted in significant changes in gene expression levels, with the increased expression of pro-apoptotic genes and decreased expression of anti-apoptotic genes. Moreover, anti-let-7a treatment was found to increase the response of A549 cells to cisplatin by reducing the expression of oncogenes akt1 and pik3ca. This study suggests that anti-let-7a treatment may enhance the A549 LC cell sensitivity to cisplatin by modulating the expression of akt1 and pik3ca genes, making it a promising therapeutic target for LC treatment.
Collapse
Affiliation(s)
- Elif Sibel Aslan
- Molecular Biology and Genetics Department, Faculty of Engineering and Natural Sciences, Biruni University, Merkezefendi, 75 Sk No:1-13 M. G, 34015, Zeytinburnu, İstanbul, Turkey.
| | - Ece Aydin
- Molecular Biology and Genetics Department, Faculty of Engineering and Natural Sciences, Biruni University, Merkezefendi, 75 Sk No:1-13 M. G, 34015, Zeytinburnu, İstanbul, Turkey
| |
Collapse
|
18
|
Zhang W, Qin H, Wang G, Zhang J, He W, Feng C, Wan H, Wang F, Guo Z. Deciphering the potential role of PGRN in regulating CD8 + T cell antitumor immunity. Cell Death Discov 2024; 10:233. [PMID: 38744851 PMCID: PMC11094002 DOI: 10.1038/s41420-024-02001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
A key factor contributing to resistance in immune checkpoint blockade (ICB) therapies is CD8+ T-cell tolerance in the tumor microenvironment (TME), partly resulting from upregulating coinhibitory receptors. Here, we describe the role of PGRN as a coinhibitory molecule that modulates the antitumor response of CD8+ T cells, thus presenting a novel immunosuppressive target for lung cancer. The in vivo subcutaneous transplanted lung cancer model showed that PGRN expression was elevated on CD8+ T cells that infiltrated transplanted lung cancers. Furthermore, PGRN deficiency was found to specifically encourage the infiltration of CD8+ T cells, enhance their proliferation, migration, and activation, and resist apoptosis, ultimately inhibiting tumor growth. This was achieved by PGRN knockout, increasing the production of T cell chemokine CCL3, which boosts the antitumor immune response induced by CD8+ T cells. Critically, the PD-L1 inhibitor exhibited a synergistic effect in enhancing the antitumor response in PGRN-/- mice. In summary, our findings highlight the significance of PGRN as a novel target for boosting CD8+ T cells antitumor immunity and its potential to overcome the resistance in ICB therapy.
Collapse
Affiliation(s)
- Wenyu Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Medical College, Tongji University, Shanghai, 200092, China
| | - Huan Qin
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Medical College, Tongji University, Shanghai, 200092, China
| | - Guosheng Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Medical College, Tongji University, Shanghai, 200092, China
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wenjuan He
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Medical College, Tongji University, Shanghai, 200092, China
| | - Chunmei Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Medical College, Tongji University, Shanghai, 200092, China
| | - Huimin Wan
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Medical College, Tongji University, Shanghai, 200092, China
| | - Feilong Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Zhongliang Guo
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
19
|
Fu J, Liu J, Zou X, Deng M, Liu G, Sun B, Guo Y, Liu D, Li Y. Transcriptome analysis of mRNA and miRNA in the development of LeiZhou goat muscles. Sci Rep 2024; 14:9858. [PMID: 38684760 PMCID: PMC11058254 DOI: 10.1038/s41598-024-60521-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024] Open
Abstract
The progression of muscle development is a pivotal aspect of animal ontogenesis, where miRNA and mRNA exert substantial influence as prominent players. It is important to understand the molecular mechanisms involved in skeletal muscle development to enhance the quality and yield of meat produced by Leizhou goats. We employed RNA sequencing (RNA-SEQ) technology to generate miRNA-mRNA profiles in Leizhou goats, capturing their developmental progression at 0, 3, and 6 months of age. A total of 977 mRNAs and 174 miRNAs were found to be differentially expressed based on our analysis. Metabolic pathways, calcium signaling pathways, and amino acid synthesis and metabolism were found to be significantly enriched among the differentially expressed mRNA in the enrichment analysis. Meanwhile, we found that among these differentially expressed mRNA, some may be related to muscle development, such as MYL10, RYR3, and CSRP3. Additionally,, we identified five muscle-specific miRNAs (miR-127-3p, miR-133a-3p, miR-193b-3p, miR-365-3p, and miR-381) that consistently exhibited high expression levels across all three stages. These miRNAs work with their target genes (FHL3, SESN1, PACSIN3, LMCD1) to regulate muscle development. Taken together, our findings suggest that several miRNAs and mRNAs are involved in regulating muscle development and cell growth in goats. By uncovering the molecular mechanisms involved in muscle growth and development, these findings contribute valuable knowledge that can inform breeding strategies aimed at enhancing meat yield and quality in Leizhou goats.
Collapse
Affiliation(s)
- Junjie Fu
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- National Local Joint Engineering Research Center of Livestock and Poultry, South China Agricultural University, Guangzhou, 510642, China
| | - Jie Liu
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
- National Local Joint Engineering Research Center of Livestock and Poultry, South China Agricultural University, Guangzhou, 510642, China
| | - Xian Zou
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Ming Deng
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- National Local Joint Engineering Research Center of Livestock and Poultry, South China Agricultural University, Guangzhou, 510642, China
| | - Guangbin Liu
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- National Local Joint Engineering Research Center of Livestock and Poultry, South China Agricultural University, Guangzhou, 510642, China
| | - Baoli Sun
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- National Local Joint Engineering Research Center of Livestock and Poultry, South China Agricultural University, Guangzhou, 510642, China
| | - Yongqing Guo
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- National Local Joint Engineering Research Center of Livestock and Poultry, South China Agricultural University, Guangzhou, 510642, China
| | - Dewu Liu
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- National Local Joint Engineering Research Center of Livestock and Poultry, South China Agricultural University, Guangzhou, 510642, China
| | - Yaokun Li
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
20
|
Ren X, Wang X, Song H, Zhang C, Yuan J, He J, Li J, Wang Z. Long non-coding RNA LINC01554 overexpression suppresses viability, migration, and invasion of liver cancer cells through regulating miR-148b-3p/EIF4E3. Heliyon 2024; 10:e27319. [PMID: 38501022 PMCID: PMC10945188 DOI: 10.1016/j.heliyon.2024.e27319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024] Open
Abstract
Background Long non-coding RNAs (lncRNAs) can be severed as competing endogenous RNAs (ceRNAs) to regulate target genes or mRNAs via sponging microRNAs (miRNAs). This study explored the effect of LINC01554 on liver cancer cells through the ceRNA mechanism. Methods Five significantly down-regulated lncRNAs were selected for further verification, and then through bioinformatics, interactive miRNAs and mRNAs of lncRNAs were identified. The relationship between LINC01554, miR-148b-3p and EIF4E3 was detected by the dual luciferase reporter gene assay. Afterwards, HCCLM3 cells were transfected with pCDH-LINC01554, miR-148b-3p inhibitor and miR-148b-3p mimics. Cell viability, apoptosis, migration and invasion were measured by Cell Counting Kit-8, flow cytometer, and Transwell assays. Real-time quantitative PCR (RT-qPCR) and Western blot were used to measure the expressions of related genes and proteins. Results LINC01554 was significantly down-regulated in the liver cancer cell lines, and was expressed in the cytoplasm of HCCLM3 cells. LINC01554 overexpression inhibited proliferation, migration, and invasion of HCCLM3 cells, and promote their apoptosis (P < 0.05). Besides, LINC01554 overexpression also significantly increased the levels of BAX, BCL2/BAX, P53, cleaved-Caspase3, TIMP3, E-cadherin and EIF4E3 (P < 0.05). Through bioinformatics and dual-luciferase reporter gene assay, LINC01554, miR-148b-3p and EIF4E3 were proved to interact with each other. Furthermore, the effects of miR-148b-3p knockdown on HCCLM3 cells were similar with those of LINC01554 overexpression, and miR-148b-3p mimics could reverse the changes of cell viability, apoptosis, migration, and invasion induced by LINC01554 overexpression. Conclusions LINC01554 overexpression could suppress the growth and metastasis of HCCLM3 cells via miR-148b-3p/EIF4E3.
Collapse
Affiliation(s)
- Xiaojing Ren
- Radiological & Environment Medicine Dept, China Institute for Radiation Protection, Taiyuan, 030032, China
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Xiaoxiao Wang
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Huangqin Song
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Chao Zhang
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Junlong Yuan
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Jiefeng He
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Jianguo Li
- Radiological & Environment Medicine Dept, China Institute for Radiation Protection, Taiyuan, 030032, China
| | - Zhuangqiang Wang
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| |
Collapse
|
21
|
Cheng X, Wu C, Xu H, Zou R, Li T, Ye S. miR-557 inhibits hepatocellular carcinoma progression through Wnt/β-catenin signaling pathway by targeting RAB10. Aging (Albany NY) 2024; 16:3716-3733. [PMID: 38364252 PMCID: PMC10929814 DOI: 10.18632/aging.205554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/26/2023] [Indexed: 02/18/2024]
Abstract
Accumulating evidence suggests that aberrant miRNAs participate in carcinogenesis and progression of hepatocellular carcinoma (HCC). Abnormal miR-557 expression is reported to interfere with the progression of several human cancers. However, the potential roles of miR-557 in HCC remain largely unknown. In the current study, we found that miR-557 was down-regulated in HCC tissues and cell lines, and was closely related to recurrence and metastasis of HCC. Notably, overexpression of miR-557 inhibited proliferation, migration, invasion, epithelial-to-mesenchymal transition (EMT) progression, blocked cells in G0/G1 phase of MHCC-97H cells in vitro, and suppressed tumor growth in vivo. However, loss of miR-557 facilitated these parameters in Huh7 cells both in vitro and in vivo. Moreover, RAB10 was identified as a direct downstream target of miR-557 through its 3'-UTR. Furthermore, RAB10 re-expression or knockdown partially abolished the effects of miR-557 on proliferation, migration, invasion, and EMT progression of HCC cells. Mechanistically, overexpression of miR-557 suppressed Wnt/β-catenin signaling by inhibiting GSK-3β phosphorylation, increasing β-catenin phosphorylation, and decreasing β-catenin transport to the nucleus, while knockdown of miR-557 activated Wnt/β-catenin signaling. Moreover, the TOP/FOP-Flash reporter assays showed that miR-557 overexpression or knockdown significantly suppressed or activated Wnt signaling activity, respectively. Additionally, low expression of miR-557 and high expression of RAB10 in HCC tissues was closely associated with tumor size, degree of differentiation, TNM stage and poor prognosis in HCC patients. Taken together, these results demonstrate that miR-557 blocks the progression of HCC via the Wnt/β-catenin pathway by targeting RAB10.
Collapse
Affiliation(s)
- Xiaoye Cheng
- Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Can Wu
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Haocheng Xu
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Ruixiang Zou
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Taiyuan Li
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Shanping Ye
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
22
|
Peng Y, Li D, Qiao B, Gao Z, Pu Q, Pang H, Lai X, Zhang R, Zhao X, Zhao G, Xu D, Han F, Wang Y, Ji Y, Pei H, Wu Q. Protonation-mediated DNA tile self-assembly with nuclease resistance characteristic for signal-amplified detection of microRNAs. Biosens Bioelectron 2024; 246:115869. [PMID: 38039736 DOI: 10.1016/j.bios.2023.115869] [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: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023]
Abstract
DNA nanotechnology, developing rapidly in recent years, has unprecedented superiorities in biological application-oriented research including high programmability, convenient functionalization, reconfigurable structure, and intrinsic biocompatibility. However, the susceptibility to nucleases in the physiological environment has been an obstacle to applying DNA nanostructures in biological science research. In this study, a new DNA self-assembly strategy, mediated by double-protonated small molecules instead of classical metal ions, is developed to enhance the nuclease resistance of DNA nanostructures while retaining their integrality and functionality, and the relative application has been launched in the detection of microRNAs (miRNAs). Faced with low-abundance miRNAs, we integrate hybrid chain reaction (HCR) with DNA self-assembly in the presence of double-protonated small molecules to construct a chemiluminescence detection platform with nuclease resistance, which utilizes the significant difference of molecular weight between DNA arrays and false-positive products to effectively separate of reaction products and remove the detection background. This strategy attaches importance to the nucleic acid stability during the assay process via improving nuclease resistance while rendering the detection results for miRNAs more authentic and reliable, opening our eyes to more possibilities for the multiple applications of customized DNA nanostructures in biology, including bioassay, bioimaging, drug delivery, and cell modulation.
Collapse
Affiliation(s)
- Yanan Peng
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, PR China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Dongxia Li
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Bin Qiao
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China; Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, 571199, PR China
| | - Zhijun Gao
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Qiumei Pu
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, PR China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Huajie Pang
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, PR China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Xiangde Lai
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, PR China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Rui Zhang
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, PR China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Xuan Zhao
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, PR China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Guangyuan Zhao
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Dan Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Pharmacy, Hainan Medical University, Haikou, 571199, PR China
| | - Feng Han
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, PR China
| | - Yuanyuan Wang
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China; Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, 571199, PR China
| | - Yuxiang Ji
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Hua Pei
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China
| | - Qiang Wu
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, PR China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, PR China; Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, 571199, PR China.
| |
Collapse
|
23
|
Luo X, Qiu Y, Fitzsimonds ZR, Wang Q, Chen Q, Lei YL. Immune escape of head and neck cancer mediated by the impaired MHC-I antigen presentation pathway. Oncogene 2024; 43:388-394. [PMID: 38177410 DOI: 10.1038/s41388-023-02912-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024]
Abstract
Tumor immune evasion is a hallmark of Head and Neck Cancers. The advent of immune checkpoint inhibitors (ICIs) in the first-line setting has transformed the management of these tumors. Unfortunately, the response rate of Head and Neck Squamous Cell Carcinomas (HNSCC) to ICIs is below 15%, regardless of the human papillomavirus (HPV) status, which might be partially related with impaired antigen presentation machinery (APM). Mechanistically, HNSCC cells are usually defective in the expression of MHC-I associated APM, while this transcriptional pathway is critical for the activation of tumor-killing effector T-cells. To specifically illuminate the phenomenon and seek for therapeutic strategies, this review summarizes the most recently identified role of genetic and functional dysregulation of the MHC-I pathway, specifically through changes at the genetic, epigenetic, post-transcriptional, and post-translational levels, which substantially contributes to HNSCC immune escape and ICI resistance. Several treatment modalities can be potentially exploited to restore APM signaling in tumors, which improves anti-tumor immunity through the activation of interferons, vaccines or rimantadine against HPV and the inhibition of EGFR, SHP-2, PI3K and MEK. Additionally, the combinatorial use of radiotherapy or cytotoxic agents with ICIs can synergize to potentiate APM signaling. Future directions would include further dissection of MHC-I related APM signaling in HNSCC and whether reversing this inhibition in combination with ICIs would elicit a more robust immune response leading to improved response rates in HNSCC. Therapeutic approaches to restore the MHC-I antigen presentation machinery in Head and Neck Cancer. (Red color texts represent the according strategies and the outcomes).
Collapse
Affiliation(s)
- Xiaobo Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Qiu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zackary R Fitzsimonds
- Department of Periodontics and Oral Medicine, Department of Otolaryngology-Head and Neck Surgery, Rogel Cancer Center, the University of Michigan, Ann Arbor, MI, 48109, USA
| | - Qiuhao Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yu Leo Lei
- Department of Periodontics and Oral Medicine, Department of Otolaryngology-Head and Neck Surgery, Rogel Cancer Center, the University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
24
|
Rodrigues P, Bangali H, Hammoud A, Mustafa YF, Al-Hetty HRAK, Alkhafaji AT, Deorari MM, Al-Taee MM, Zabibah RS, Alsalamy A. COX 2-inhibitors; a thorough and updated survey into combinational therapies in cancers. Med Oncol 2024; 41:41. [PMID: 38165473 DOI: 10.1007/s12032-023-02256-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024]
Abstract
Cyclooxygenase (COX) enzymes are pivotal in inflammation and cancer development. COX-2, in particular, has been implicated in tumor growth, angiogenesis, and immune evasion. Recently, COX-2 inhibitors have arisen as potential therapeutic agents in cancer treatment. In addition, combining COX inhibitors with other treatment modalities has demonstrated the potential to improve therapeutic efficacy. This review aims to investigate the effects of COX inhibition, both alone and in combination with other methods, on signaling pathways and carcinogenesis in various cancers. In this study, a literature search of all major academic databases was conducted (PubMed, Scholar google), including the leading research on the mechanisms of COX-2, COX-2 inhibitors, monotherapy with COX-2 inhibitors, and combining COX-2-inhibitors with chemotherapeutic agents in tumors. The study encompasses preclinical and clinical evidence, highlighting the positive findings and the potential implications for clinical practice. According to preclinical studies, multiple signaling pathways implicated in tumor cell proliferation, survival, invasion, and metastasis can be suppressed by inhibiting COX. In addition, combining COX inhibitors with chemotherapy drugs, targeted therapies, immunotherapies, and miRNA-based approaches has enhanced anti-tumor activity. These results suggest that combination therapy has the potential to overcome resistance mechanisms and improve treatment outcomes. However, caution must be exercised when selecting and administering combination regimens. Not all combinations of COX-2 inhibitors with other drugs result in synergistic effects; some may even have unfavorable interactions. Therefore, personalized approaches that consider the specific characteristics of the cancer and the medications involved are crucial for optimizing therapeutic strategies. In conclusion, as monotherapy or combined with other methods, COX inhibition bears promise in modulating signaling pathways and inhibiting carcinogenesis in various cancers. Additional studies and well-designed clinical trials are required to completely elucidate the efficacy of COX inhibition and combination therapy in enhancing cancer treatment outcomes. This narrative review study provides a detailed summary of COX-2 monotherapy and combination targeted therapy in cancer treatment.
Collapse
Affiliation(s)
- Paul Rodrigues
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Asir-Abha, Kingdom of Saudi Arabia
| | - Harun Bangali
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Asir-Abha, Kingdom of Saudi Arabia
| | - Ahmad Hammoud
- Department of Medical and Technical Information Technology, Bauman Moscow State Technical University, Moscow, Russia.
- Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, Mishref Campus, Mubarak Al-Abdullah, Kuwait.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | | | | - Maha Medha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Rahman S Zabibah
- College of Medical Technique, the Islamic University, Najaf, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, 66002, Iraq
| |
Collapse
|
25
|
Sharma S, Rana R, Prakash P, Ganguly NK. Drug target therapy and emerging clinical relevance of exosomes in meningeal tumors. Mol Cell Biochem 2024; 479:127-170. [PMID: 37016182 PMCID: PMC10072821 DOI: 10.1007/s11010-023-04715-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/17/2023] [Indexed: 04/06/2023]
Abstract
Meningioma is the most common central nervous system (CNS) tumor. In recent decades, several efforts have been made to eradicate this disease. Surgery and radiotherapy remain the standard treatment options for these tumors. Drug therapy comes to play its role when both surgery and radiotherapy fail to treat the tumor. This mostly happens when the tumors are close to vital brain structures and are nonbenign. Although a wide variety of chemotherapeutic drugs and molecular targeted drugs such as tyrosine kinase inhibitors, alkylating agents, endocrine drugs, interferon, and targeted molecular pathway inhibitors have been studied, the roles of numerous drugs remain unexplored. Recent interest is growing toward studying and engineering exosomes for the treatment of different types of cancer including meningioma. The latest studies have shown the involvement of exosomes in the theragnostic of various cancers such as the lung and pancreas in the form of biomarkers, drug delivery vehicles, and vaccines. Proper attention to this new emerging technology can be a boon in finding the consistent treatment of meningioma.
Collapse
Affiliation(s)
- Swati Sharma
- Department of Research, Sir Ganga Ram Hospital, New Delhi, 110060 India
| | - Rashmi Rana
- Department of Research, Sir Ganga Ram Hospital, New Delhi, 110060 India
| | - Prem Prakash
- Department of Molecular Medicine, Jamia Hamdard, New Delhi, 110062 India
| | | |
Collapse
|
26
|
McWhorter R, Bonavida B. The Role of TAMs in the Regulation of Tumor Cell Resistance to Chemotherapy. Crit Rev Oncog 2024; 29:97-125. [PMID: 38989740 DOI: 10.1615/critrevoncog.2024053667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Tumor-associated macrophages (TAMs) are the predominant cell infiltrate in the immunosuppressive tumor microenvironment (TME). TAMs are central to fostering pro-inflammatory conditions, tumor growth, metastasis, and inhibiting therapy responses. Many cancer patients are innately refractory to chemotherapy and or develop resistance following initial treatments. There is a clinical correlation between the level of TAMs in the TME and chemoresistance. Hence, the pivotal role of TAMs in contributing to chemoresistance has garnered significant attention toward targeting TAMs to reverse this resistance. A prerequisite for such an approach requires a thorough understanding of the various underlying mechanisms by which TAMs inhibit response to chemotherapeutic drugs. Such mechanisms include enhancing drug efflux, regulating drug metabolism and detoxification, supporting cancer stem cell (CSCs) resistance, promoting epithelial-mesenchymal transition (EMT), inhibiting drug penetration and its metabolism, stimulating angiogenesis, impacting inhibitory STAT3/NF-κB survival pathways, and releasing specific inhibitory cytokines including TGF-β and IL-10. Accordingly, several strategies have been developed to overcome TAM-modulated chemoresistance. These include novel therapies that aim to deplete TAMs, repolarize them toward the anti-tumor M1-like phenotype, or block recruitment of monocytes into the TME. Current results from TAM-targeted treatments have been unimpressive; however, the use of TAM-targeted therapies in combination appears promising These include targeting TAMs with radiotherapy, chemotherapy, chemokine receptor inhibitors, immunotherapy, and loaded nanoparticles. The clinical limitations of these strategies are discussed.
Collapse
Affiliation(s)
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
27
|
Chen R, Coleborn E, Bhavsar C, Wang Y, Alim L, Wilkinson AN, Tran MA, Irgam G, Atluri S, Wong K, Shim JJ, Adityan S, Lee JS, Overwijk WW, Steptoe R, Yang D, Wu SY. miR-146a inhibits ovarian tumor growth in vivo via targeting immunosuppressive neutrophils and enhancing CD8 + T cell infiltration. Mol Ther Oncolytics 2023; 31:100725. [PMID: 37781339 PMCID: PMC10539880 DOI: 10.1016/j.omto.2023.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/08/2023] [Indexed: 10/03/2023] Open
Abstract
Immunotherapies have emerged as promising strategies for cancer treatment. However, existing immunotherapies have poor activity in high-grade serous ovarian cancer (HGSC) due to the immunosuppressive tumor microenvironment and the associated low tumoral CD8+ T cell (CTL) infiltration. Through multiple lines of evidence, including integrative analyses of human HGSC tumors, we have identified miR-146a as a master regulator of CTL infiltration in HGSC. Tumoral miR-146a expression is positively correlated with anti-cancer immune signatures in human HGSC tumors, and delivery of miR-146a to tumors resulted in significant reduction in tumor growth in both ID8-p53-/- and IG10 murine HGSC models. Increasing miR-146a expression in tumors improved anti-tumor immune responses by decreasing immune suppressive neutrophils and increasing CTL infiltration. Mechanistically, miR-146a targets IL-1 receptor-associated kinase 1 and tumor necrosis factor receptor-associated factor 6 adaptor molecules of the transcription factor nuclear factor κB signaling pathway in ID8-p53-/- cells and decreases production of the downstream neutrophil chemoattractant, C-X-C motif chemokine ligand 1. In addition to HGSC, tumoral miR-146a expression also correlates strongly with CTL infiltration in other cancer types including thyroid, prostate, breast, and adrenocortical cancers. Altogether, our findings highlight the ability of miR-146a to overcome immune suppression and improve CTL infiltration in tumors.
Collapse
Affiliation(s)
- Rui Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Elaina Coleborn
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Chintan Bhavsar
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yue Wang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Louisa Alim
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Andrew N. Wilkinson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | | | - Gowri Irgam
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sharat Atluri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kiefer Wong
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jae-Jun Shim
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Siddharth Adityan
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Willem W. Overwijk
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Raymond Steptoe
- Frazer Institute, University of Queensland, Brisbane, QLD 4102, Australia
| | - Da Yang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Sherry Y. Wu
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
28
|
Wang C, Bai M, Sun Z, Yao N, Zhang A, Guo S, Asemi Z. Epigallocatechin-3-gallate and cancer: focus on the role of microRNAs. Cancer Cell Int 2023; 23:241. [PMID: 37838685 PMCID: PMC10576883 DOI: 10.1186/s12935-023-03081-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/21/2023] [Indexed: 10/16/2023] Open
Abstract
MicroRNAs (miRNAs) are a group of small non-coding RNAs that affect gene expression. The role of miRNAs in different types of cancers has been published and it was shown that several miRNAs are inappropriately expressed in different cancers. Among the mechanisms that can cause this lack of proper expression are epigenetics, chromosomal changes, polymorphisms or defects in processing proteins. Recent research shows that phytochemicals, including epigallocatechin-3-gallate (EGCG), exert important epigenetic-based anticancer effects such as pro-apoptotic or anti proliferative through miRNA gene silencing. Given that EGCG is able to modulate a variety of cancer-related process i.e., angiogenesis, proliferation, metastasis and apoptosis via targeting various miRNAs such as let-7, miR-16, and miR-210. The discovery of new miRNAs and the differences observed in their expression when exposed to EGCG provides evidence that targeting these miRNAs may be beneficial as a form of treatment. In this review, we aim to provide an overview, based on current knowledge, on how phytochemicals, including epigallocatechin-3-gallate, can be considered as potential miRNAs modulator to improve efficacy of current cancer treatments.
Collapse
Affiliation(s)
- Chunguang Wang
- The First Affiliated Hospital of Hebei North University, Zhang Jiakou, 075000, Hebei, China
| | - Meiling Bai
- Basic Medical College of Hebei North University, Zhang Jiakou, 075000, Hebei, China.
| | - Zhiguang Sun
- The First Affiliated Hospital of Hebei North University, Zhang Jiakou, 075000, Hebei, China
| | - Nan Yao
- The First Affiliated Hospital of Hebei North University, Zhang Jiakou, 075000, Hebei, China
| | - Aiting Zhang
- The First Affiliated Hospital of Hebei North University, Zhang Jiakou, 075000, Hebei, China
| | - Shengyu Guo
- The First Affiliated Hospital of Hebei North University, Zhang Jiakou, 075000, Hebei, China
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
29
|
Chamoto K, Yaguchi T, Tajima M, Honjo T. Insights from a 30-year journey: function, regulation and therapeutic modulation of PD1. Nat Rev Immunol 2023; 23:682-695. [PMID: 37185300 DOI: 10.1038/s41577-023-00867-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 05/17/2023]
Abstract
PD1 was originally discovered in 1992 as a molecule associated with activation-induced cell death in T cells. Over the past 30 years, it was found that PD1 has a critical role in avoiding overactivation-induced cell death and autoimmunity, whereas its inhibition unleashes anticancer immunity. Here, we outline the journey from the discovery of PD1 to its role as a breakthrough target in cancer immunotherapy. We describe its regulation and function and examine how a mechanistic understanding of PD1 signalling suggests a central function in setting the T cell activation threshold, thereby controlling T cell proliferation, differentiation, exhaustion and metabolic status. This threshold theory, in combination with new insights into T cell metabolism and a better understanding of immune cell modulation by the microbiota, can provide guidance for the development of efficient combination therapies. Moreover, we discuss the mechanisms underlying immune-related adverse events after PD1-targeted therapy and their possible treatment.
Collapse
Affiliation(s)
- Kenji Chamoto
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomonori Yaguchi
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Tajima
- Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tasuku Honjo
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
30
|
Zhang Y, Zheng J, Li F, Ge C, Zhang H. MicroRNA transcriptome analysis for elucidating the immune mechanism of the redclaw crayfish Cherax quadricarinatus under Decapod iridescent virus 1 infection. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109026. [PMID: 37633343 DOI: 10.1016/j.fsi.2023.109026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Redclaw crayfish (Cherax quadricarinatus) is a large, tropical freshwater crustacean species with considerable potential of commercial production. In recent years, infection with DIV1 in redclaw crayfish is being reported in aquaculture industries, causing high mortality and huge economic losses. However, many characteristics of this virus, including pathogenesis, transmission mechanism, and host immunity, remain largely unknown.MicroRNAs are known to play important roles in numerous biological processes, and many microRNAs are reported to be involved in the regulation of immune responses. In this study, nine-small RNA libraries were constructed using hemocytes of redclaw crayfish to characterize the differentially expressed miRNAs (DE-miRNAs) at 24 and 48 h postinfection (hpi). A total of 14 and 22 DE-miRNAs were identified in response to DIV1 infection at 24 and 48 hpi, respectively. Further, functional annotation of the predicted host target genes using GO and KEGG pathway enrichment analyses indicated that relevant biological processes and signal pathways underwent miRNA-mediated regulation after DIV1 infection. Our results enhanced the understanding of the mechanisms of miRNA-mediated regulation of immune responses under DIV1 infection in crustaceans.
Collapse
Affiliation(s)
- Yucheng Zhang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, China; Key Laboratory of Genetics and Breeding, Zhejiang Institute of Freshwater Fisheries, Huzhou, China
| | - Jianbo Zheng
- Key Laboratory of Genetics and Breeding, Zhejiang Institute of Freshwater Fisheries, Huzhou, China
| | - Fei Li
- Key Laboratory of Genetics and Breeding, Zhejiang Institute of Freshwater Fisheries, Huzhou, China
| | - Chutian Ge
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, China
| | - Haiqi Zhang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, China; Key Laboratory of Genetics and Breeding, Zhejiang Institute of Freshwater Fisheries, Huzhou, China.
| |
Collapse
|
31
|
Morgunova A, Ibrahim P, Chen GG, Coury SM, Turecki G, Meaney MJ, Gifuni A, Gotlib IH, Nagy C, Ho TC, Flores C. Preparation and processing of dried blood spots for microRNA sequencing. Biol Methods Protoc 2023; 8:bpad020. [PMID: 37901452 PMCID: PMC10603595 DOI: 10.1093/biomethods/bpad020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Dried blood spots (DBS) are biological samples commonly collected from newborns and in geographic areas distanced from laboratory settings for the purposes of disease testing and identification. MicroRNAs (miRNAs)-small non-coding RNAs that regulate gene activity at the post-transcriptional level-are emerging as critical markers and mediators of disease, including cancer, infectious diseases, and mental disorders. This protocol describes optimized procedural steps for utilizing DBS as a reliable source of biological material for obtaining peripheral miRNA expression profiles. We outline key practices, such as the method of DBS rehydration that maximizes RNA extraction yield, and the use of degenerate oligonucleotide adapters to mitigate ligase-dependent biases that are associated with small RNA sequencing. The standardization of miRNA readout from DBS offers numerous benefits: cost-effectiveness in sample collection and processing, enhanced reliability and consistency of miRNA profiling, and minimal invasiveness that facilitates repeated testing and retention of participants. The use of DBS-based miRNA sequencing is a promising method to investigate disease mechanisms and to advance personalized medicine.
Collapse
Affiliation(s)
- Alice Morgunova
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Pascal Ibrahim
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec H3A 0G4, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec H4H 1R3, Canada
| | - Gary Gang Chen
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec H3A 0G4, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec H4H 1R3, Canada
| | - Saché M Coury
- Department of Psychology, Stanford University, Stanford, CA 94305, United States
- Department of Psychology, University of California, Los Angeles, CA 90095, United States
| | - Gustavo Turecki
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec H3A 0G4, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Michael J Meaney
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 0G4, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec H3A 2B4, Canada
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, Singapore City 138632, Singapore
| | - Anthony Gifuni
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Ian H Gotlib
- Department of Psychology, Stanford University, Stanford, CA 94305, United States
| | - Corina Nagy
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec H3A 0G4, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Tiffany C Ho
- Department of Psychology, Stanford University, Stanford, CA 94305, United States
- Department of Psychology, University of California, Los Angeles, CA 90095, United States
| | - Cecilia Flores
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 0G4, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
32
|
Al-Asadi S, Mansour H, Ataimish AJ, Al-Kahachi R, Rampurawala J. MicroRNAs Regulate Tumorigenesis by Downregulating SOCS3 Expression: An In silico Approach. Bioinform Biol Insights 2023; 17:11779322231193535. [PMID: 37701630 PMCID: PMC10493049 DOI: 10.1177/11779322231193535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 07/24/2023] [Indexed: 09/14/2023] Open
Abstract
Tumor microenvironment is characterized by the occurrence of significant changes due to disrupted signaling pathways that affect a broad spectrum of cellular activities such as proliferation, differentiation, signaling, invasiveness, migration, and apoptosis. Similarly, a downregulated suppressor of cytokine signaling 3 (SOCS3) promotes increased JAK/STAT function due to aberrant cytokine signaling, which results in increased cell proliferation, differentiation, and migration. Multiple carcinomas including breast cancer, prostate cancer, hepatocellular carcinoma, pancreatic cancer, and colorectal cancer involve the disruption of SOCS3 expression due to microRNA overexpression. MicroRNAs are small, conserved, and non-coding RNA molecules that regulate gene expression through post-transcriptional inhibition and mRNA destabilization. The aim of this study was to identify putative microRNAs that interact with SOCS3 and downregulate its expression. In this study, miRWalk, TargetScan, and miRDB were used to identify microRNAs that interact with SOCS3, whereas RNA22 was utilized to identify the binding sites of 238 significant microRNAs. The tertiary structures of shortlisted microRNAs and SOCS3 regions were predicted through MC Sym and RNAComposer, respectively. For molecular docking, HDOCK was used, which predicted 80 microRNA-messengerRNA complexes and the interactions of the top 5 shortlisted complexes were assessed. The complexes were shortlisted on the basis of least binding affinity score and maximum confidence score. This study identifies the interactions of known (miR-203a-5p) and novel (miR-6756-5p, miR-6732-5p, miR-1203, miR-6887-5p) microRNAs with SOCS3 regions due to their maximum interactions. Identifying the interactions of these microRNAs with SOCS3 will significantly advance the understanding of oncomiRs (miRNAs that are associated with cancer development) in tumor development due to their influence on SOCS3 expression. These insights will assist in future studies to understand the significance of miRNA-SOCS3-associated tumor development and progression.
Collapse
Affiliation(s)
- Sura Al-Asadi
- College of Health and Medical Techniques, Middle Technical University, Baghdad, Iraq
| | - Hiba Mansour
- College of Health and Medical Techniques, Middle Technical University, Baghdad, Iraq
| | | | - Rusul Al-Kahachi
- Department of Scholarships and Cultural Relationship, Republic of Iraq Ministry of Higher Education and Scientific Research, Baghdad, Iraq
| | | |
Collapse
|
33
|
He Z, Zhu Q. Circular RNAs: Emerging roles and new insights in human cancers. Biomed Pharmacother 2023; 165:115217. [PMID: 37506578 DOI: 10.1016/j.biopha.2023.115217] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Circular RNAs (circRNAs) are single-stranded, covalently closed RNA molecules formed by mRNA exon back-splicing. Although the circRNA functions remain largely unknown, their currently known biological activities include: acting as competing endogenous RNA (ceRNA) to adsorb microRNA (miRNA), binding proteins, regulating transcription or splicing, and ability to be translated into proteins or peptides. A growing number of studies have found that many circRNAs are abnormally expressed in various cancers, and their dysregulation is highly correlated with tumor progression. Therefore, diagnosis and treatment using circRNAs as biomarkers and therapeutic targets, respectively, has gradually become an attractive research topic. In this review, we introduced the canonical biogenesis pathways and degradation mechanisms of circRNAs. In addition, we examined the biological functions of circRNAs in vivo. Finally, we discussed the current clinical applications and challenges faced by circRNA, and proposed future directions for this promising research field.
Collapse
Affiliation(s)
- Zhilin He
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha 410013, Hunan, China.
| | - Qubo Zhu
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
34
|
Liu K, He S, Sun S, Zhang X, He Y, Quan F, Pang B, Xiao Y. Computational Quantification of Cancer Immunoediting. Cancer Immunol Res 2023; 11:1159-1167. [PMID: 37540180 DOI: 10.1158/2326-6066.cir-22-0926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/31/2023] [Accepted: 07/10/2023] [Indexed: 08/05/2023]
Abstract
The remarkable success of cancer immunotherapy has revolutionized cancer treatment, emphasizing the importance of tumor-immune interactions in cancer evolution and treatment. Cancer immunoediting describes the dual effect of tumor-immune interactions: inhibiting tumor growth by destroying tumor cells and facilitating tumor escape by shaping tumor immunogenicity. To better understand tumor-immune interactions, it is critical to develop computational methods to measure the extent of cancer immunoediting. In this review, we provide a comprehensive overview of the computational methods for quantifying cancer immunoediting. We focus on describing the basic ideas, computational processes, advantages, limitations, and influential factors. We also summarize recent advances in quantifying cancer immunoediting studies and highlight future research directions. As the methods for quantifying cancer immunoediting are continuously improved, future research will further help define the role of immunity in tumorigenesis and hopefully provide a basis for the design of new personalized cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Kun Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shengyuan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shangqin Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanzhen He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Fei Quan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Bo Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
35
|
Yang K, Halima A, Chan TA. Antigen presentation in cancer - mechanisms and clinical implications for immunotherapy. Nat Rev Clin Oncol 2023; 20:604-623. [PMID: 37328642 DOI: 10.1038/s41571-023-00789-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/18/2023]
Abstract
Over the past decade, the emergence of effective immunotherapies has revolutionized the clinical management of many types of cancers. However, long-term durable tumour control is only achieved in a fraction of patients who receive these therapies. Understanding the mechanisms underlying clinical response and resistance to treatment is therefore essential to expanding the level of clinical benefit obtained from immunotherapies. In this Review, we describe the molecular mechanisms of antigen processing and presentation in tumours and their clinical consequences. We examine how various aspects of the antigen-presentation machinery (APM) shape tumour immunity. In particular, we discuss genomic variants in HLA alleles and other APM components, highlighting their influence on the immunopeptidomes of both malignant cells and immune cells. Understanding the APM, how it is regulated and how it changes in tumour cells is crucial for determining which patients will respond to immunotherapy and why some patients develop resistance. We focus on recently discovered molecular and genomic alterations that drive the clinical outcomes of patients receiving immune-checkpoint inhibitors. An improved understanding of how these variables mediate tumour-immune interactions is expected to guide the more precise administration of immunotherapies and reveal potentially promising directions for the development of new immunotherapeutic approaches.
Collapse
Affiliation(s)
- Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Ahmed Halima
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA.
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA.
- National Center for Regenerative Medicine, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
36
|
Zhang H, Cao K, Xiang J, Zhang M, Zhu M, Xi Q. Hypoxia induces immunosuppression, metastasis and drug resistance in pancreatic cancers. Cancer Lett 2023; 571:216345. [PMID: 37558084 DOI: 10.1016/j.canlet.2023.216345] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023]
Abstract
Pancreatic cancer is one of the common malignant tumors of the digestive system and is known as the "king of cancers". It is extremely difficult to diagnose at an early stage, the disease progresses rapidly, and the effect of chemotherapy and radiotherapy is poor, so the prognosis of pancreatic cancer patients is very poor. Numerous studies have suggested that hypoxia is closely related to the development and progression of pancreatic cancer. Inadequate blood supply and desmoplasia in the microenvironment of pancreatic cancer can result in its extreme hypoxia. This hypoxic microenvironment can further contribute to angiogenesis and desmoplasia. Hypoxia is mediated by the complex hypoxia inducible factor (HIF) signaling pathway and plays an important role in the formation of a highly immunosuppressive microenvironment and the metastasis of pancreatic cancer. Further work on the hypoxic microenvironment will help clarify the specific mechanisms of the role of hypoxia in pancreatic cancer and provide a basis for the realization of hypoxia-targeted therapeutic and diagnostic strategies.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Kailei Cao
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Jingrong Xiang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Mengting Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Mengxin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Qinhua Xi
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
37
|
Sari G, Rock KL. Tumor immune evasion through loss of MHC class-I antigen presentation. Curr Opin Immunol 2023; 83:102329. [PMID: 37130455 PMCID: PMC10524158 DOI: 10.1016/j.coi.2023.102329] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 05/04/2023]
Abstract
CD8 T cells recognize cancers when they detect antigenic peptides presented on a tumor's surface MHC-I molecules. Since MHC-I antigen presentation is not essential for cell growth or survival, many cancers inactivate this pathway, and thereby escape control by CD8 T cells. Such immune evasion allows cancers to progress and also become resistant to CD8 T- cell-based immunotherapies, such as checkpoint blockade. Here, we review recent findings about the various different mechanisms that cancers use to impair antigen presentation, the consequence of such changes, and, in some cases, the potential to reverse these defects.
Collapse
Affiliation(s)
- Gulce Sari
- University of Massachusetts Medical School, Department of Pathology, Worcester, MA, USA
| | - Kenneth L Rock
- University of Massachusetts Medical School, Department of Pathology, Worcester, MA, USA.
| |
Collapse
|
38
|
Bogaczyk A, Zawlik I, Zuzak T, Kluz M, Potocka N, Kluz T. The Role of miRNAs in the Development, Proliferation, and Progression of Endometrial Cancer. Int J Mol Sci 2023; 24:11489. [PMID: 37511248 PMCID: PMC10380838 DOI: 10.3390/ijms241411489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/30/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Endometrial cancer is one of the most common cancers in developing and developed countries. Although the detection of this cancer is high at the early stages, there is still a lack of markers to monitor the disease, its recurrence, and metastasis. MiRNAs are in charge of the post-transcriptional regulation of genes responsible for the most important biological processes, which is why they are increasingly used as biomarkers in many types of cancer. Many studies have demonstrated the influence of miRNAs on the processes related to carcinogenesis. The characteristics of miRNA expression profiles in endometrial cancer will allow their use as diagnostic and prognostic biomarkers. This paper focuses on the discussion of selected miRNAs based on the literature and their role in the development of endometrial cancer.
Collapse
Affiliation(s)
- Anna Bogaczyk
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, F.Szopena 2, 35-055 Rzeszow, Poland; (A.B.); (T.Z.); (T.K.)
| | - Izabela Zawlik
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, Warzywna 1a, 35-959 Rzeszow, Poland;
- Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland
| | - Tomasz Zuzak
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, F.Szopena 2, 35-055 Rzeszow, Poland; (A.B.); (T.Z.); (T.K.)
| | - Marta Kluz
- Department of Pathology, Fryderyk Chopin University Hospital, F.Szopena 2, 35-055 Rzeszow, Poland;
| | - Natalia Potocka
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, Warzywna 1a, 35-959 Rzeszow, Poland;
| | - Tomasz Kluz
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, F.Szopena 2, 35-055 Rzeszow, Poland; (A.B.); (T.Z.); (T.K.)
- Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland
| |
Collapse
|
39
|
Hu R, Zhang W, Han Z, Ma M, Huang Q, Lv M, Ma W, Sun X, Feng W, Li J, Zhong X, Sun J, Yao W, Zhou X. Identification of immune-related target and prognostic biomarkers in PBMC of hepatocellular carcinoma. BMC Gastroenterol 2023; 23:234. [PMID: 37438735 DOI: 10.1186/s12876-023-02843-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 05/31/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide, and is characterized by insidious onset, rapid progression, and poor prognosis. Immunotherapy is a first-line treatment for advanced HCC. The identification of immune-related prognostic markers may be an effective strategy to predict and improve clinical response rate of immunotherapy. METHODS The DESeq2, edgeR, and limma R packages were used to compare the transcriptomes of HCC with different prognoses. Cancer-related databases such as UALCAN, TNMplot, GEPIA, muttarget and Human Protein Atlas (HPA), and the Kaplan-Meier Plotter platform were used to analyze the relationship between CLDN18 and the clinical characteristics, as well as prognosis of HCC. The co-expressed genes of CLDN18 were obtained from LinkedOmics platform, and GO functional enrichment and KEGG pathway analysis were performed. The CIBERSORT, TIMER, Timer 2.0 and TISIDB algorithms were used to analyze immune infiltration. RESULTS CLDN18 was differentially expressed in HCC patients with different prognoses, and its expression level in PBMC was positively correlated with the stage of BCLC. In addition, CLDN18 was significantly overexpressed in HCC tumor tissues compared to adjacent non-tumor tissues, which was consistent with PBMC sequencing results and immunohistochemical data from human protein profiles. CLDN18 was also positively correlated with HCC staging and grading, and high expression levels of CLDN18 predicted shorter overall survival. Functional annotation of CLDN18 in HCC revealed enrichment of the cellular senescence and protein activation cascade, along with biological processes such as cell cycle, inflammatory response, and cellular ketone metabolism. In addition, CLDN18 was also associated with tumor infiltrating immune cells, suppressive immune cell markers, T lymphocyte depletion and activation of HCC, and low expression of CLDN18 was associated with higher CD8 + T cell infiltration and better survival rates. CONCLUSIONS CLDN18 is a potential prognostic marker and immunotherapeutic target for HCC.
Collapse
Affiliation(s)
- Rui Hu
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Wei Zhang
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
| | - Zhiyi Han
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
| | - Mengqing Ma
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Qi Huang
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Minling Lv
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Wenfeng Ma
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
| | - Xinfeng Sun
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
| | - Wenxing Feng
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
| | - Jing Li
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
| | - Xin Zhong
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Jialing Sun
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
| | - Wei Yao
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China
| | - Xiaozhou Zhou
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Futian District, Shenzhen, 518033, Guangdong Province, China.
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China.
| |
Collapse
|
40
|
Bao L, Zhu P, Mou Y, Song Y, Qin Y. Targeting LSD1 in tumor immunotherapy: rationale, challenges and potential. Front Immunol 2023; 14:1214675. [PMID: 37483603 PMCID: PMC10360200 DOI: 10.3389/fimmu.2023.1214675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Lysine-specific demethylase 1 (LSD1) is an enzyme that removes lysine methylation marks from nucleosome histone tails and plays an important role in cancer initiation, progression, metastasis, and recurrence. Recent research shows that LSD1 regulates tumor cells and immune cells through multiple upstream and downstream pathways, enabling tumor cells to adapt to the tumor microenvironment (TME). As a potential anti-tumor treatment strategy, immunotherapy has developed rapidly in the past few years. However, most patients have a low response rate to available immune checkpoint inhibitors (ICIs), including anti-PD-(L)1 therapy and CAR-T cell therapy, due to a broad array of immunosuppressive mechanisms. Notably, inhibition of LSD1 turns "cold tumors" into "hot tumors" and subsequently enhances tumor cell sensitivity to ICIs. This review focuses on recent advances in LSD1 and tumor immunity and discusses a potential therapeutic strategy for combining LSD1 inhibition with immunotherapy.
Collapse
Affiliation(s)
- Lei Bao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Ping Zhu
- Department of Nephrology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
| | - Yuan Mou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Yinhong Song
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- College of Basic Medical Science, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
| | - Ye Qin
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- College of Basic Medical Science, China Three Gorges University, Yichang, China
| |
Collapse
|
41
|
Han L, Chen S, Luan Z, Fan M, Wang Y, Sun G, Dai G. Immune function of colon cancer associated miRNA and target genes. Front Immunol 2023; 14:1203070. [PMID: 37465677 PMCID: PMC10351377 DOI: 10.3389/fimmu.2023.1203070] [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: 04/10/2023] [Accepted: 05/15/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction Colon cancer is a complex disease that involves intricate interactions between cancer cells and theimmune microenvironment. MicroRNAs (miRNAs) have recently emerged as critical regulators of gene expression in cancer, including colon cancer. There is increasing evidence suggesting that miRNA dysregulation plays a crucial role in modulating the immune microenvironment of intestinal cancer. In particular, miRNAs regulate immune cell activation, differentiation, and function, as well as cytokine and chemokine production in intestinal cancer. It is urgent to fully investigate the potential role of intestinal cancer-related miRNAs in shaping the immune microenvironment. Methods Therefore, this paper aims to identify miRNAs that are potentially associated with colon cancer and regulate a large number of genes related to immune function. We explored the role of these genes in colon cancer patient prognosis, immune infiltration, and tumor purity based on data of 174 colon cancer patients though convolutional neural network, survival analysis and multiple analysis tools. Results Our findings suggest that miRNA regulated genes play important roles in CD4 memory resting cells, macrophages.M2, and Mast cell activated cells, and they are concentrated in the cytokinecytokine receptor interaction pathway. Discussion Our study enhances our understanding of the underlying mechanisms of intestinal cancer and provides new insights into the development of effective therapies. Additionally, identification of miRNA biomarkers could aid in diagnosis and prognosis, as well as guide personalized treatment strategies for patients with intestinal cancer.
Collapse
Affiliation(s)
- Lu Han
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Shiyun Chen
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Zhe Luan
- Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Mengjiao Fan
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Yanrong Wang
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Gang Sun
- Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guanghai Dai
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
42
|
Peng Y, Gao Z, Qiao B, Li D, Pang H, Lai X, Pu Q, Zhang R, Zhao X, Zhao G, Xu D, Wang Y, Ji Y, Pei H, Wu Q. Size-Controlled DNA Tile Self-Assembly Nanostructures Through Caveolae-Mediated Endocytosis for Signal-Amplified Imaging of MicroRNAs in Living Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300614. [PMID: 37189216 PMCID: PMC10375201 DOI: 10.1002/advs.202300614] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/30/2023] [Indexed: 05/17/2023]
Abstract
Signal-amplified imaging of microRNAs (miRNAs) is a promising strategy at the single-cell level because liquid biopsy fails to reflect real-time dynamic miRNA levels. However, the internalization pathways for available conventional vectors predominantly involve endo-lysosomes, showing nonideal cytoplasmic delivery efficiency. In this study, size-controlled 9-tile nanoarrays are designed and constructed by integrating catalytic hairpin assembly (CHA) with DNA tile self-assembly technology to achieve caveolae-mediated endocytosis for the amplified imaging of miRNAs in a complex intracellular environment. Compared with classical CHA, the 9-tile nanoarrays possess high sensitivity and specificity for miRNAs, achieve excellent internalization efficiency by caveolar endocytosis, bypassing lysosomal traps, and exhibit more powerful signal-amplified imaging of intracellular miRNAs. Because of their excellent safety, physiological stability, and highly efficient cytoplasmic delivery, the 9-tile nanoarrays can realize real-time amplified monitoring of miRNAs in various tumor and identical cells of different periods, and imaging effects are consistent with the actual expression levels of miRNAs, ultimately demonstrating their feasibility and capacity. This strategy provides a high-potential delivery pathway for cell imaging and targeted delivery, simultaneously offering a meaningful reference for the application of DNA tile self-assembly technology in relevant fundamental research and medical diagnostics.
Collapse
Affiliation(s)
- Yanan Peng
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Zhijun Gao
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Bin Qiao
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
- Key Laboratory of Emergency and Trauma of Ministry of EducationResearch Unit of Island Emergency MedicineChinese Academy of Medical Sciences (No. 2019RU013)Hainan Medical UniversityHaikou571199P. R. China
| | - Dongxia Li
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Huajie Pang
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Xiangde Lai
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Qiumei Pu
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Rui Zhang
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Xuan Zhao
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Guangyuan Zhao
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Dan Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of EducationSchool of PharmacyHainan Medical UniversityHaikou571199P. R. China
| | - Yuanyuan Wang
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
- Key Laboratory of Emergency and Trauma of Ministry of EducationResearch Unit of Island Emergency MedicineChinese Academy of Medical Sciences (No. 2019RU013)Hainan Medical UniversityHaikou571199P. R. China
| | - Yuxiang Ji
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Hua Pei
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
| | - Qiang Wu
- The Second Affiliated HospitalSchool of Tropical MedicineHainan Medical UniversityHaikou571199P. R. China
- Key Laboratory of Emergency and Trauma of Ministry of EducationResearch Unit of Island Emergency MedicineChinese Academy of Medical Sciences (No. 2019RU013)Hainan Medical UniversityHaikou571199P. R. China
| |
Collapse
|
43
|
Wu YY, Wang Q, Zhang PA, Zhu C, Xu GY. miR-1306-3p directly activates P2X3 receptors in primary sensory neurons to induce visceral pain in rats. Pain 2023; 164:1555-1565. [PMID: 36633528 PMCID: PMC10281022 DOI: 10.1097/j.pain.0000000000002853] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/23/2022] [Accepted: 11/08/2022] [Indexed: 01/13/2023]
Abstract
ABSTRACT Mounting evidence indicates that microRNAs (miRNAs) play critical roles in various pathophysiological conditions and diseases, but the physiological roles of extracellular miRNAs on the disease-related ion channels remain largely unknown. Here, we showed that miR-1306-3p evoked action potentials and induced inward currents of the acutely isolated rat dorsal root ganglion (DRG) neurons. The miR-1306-3p-induced effects were significantly inhibited by A317491, a potent inhibitor of the P2X3 receptor (P2X3R), or disappeared after the knockdown of P2X3Rs in DRG neurons. We further identified R180, K315, and R52 as the miR-1306-3p interaction sites on the extracellular domain of P2X3Rs, which were distinct from the orthosteric ATP-binding sites. Intrathecal injection of miR-1306-3p produced visceral pain but not somatic pain in normal control rats. Conversely, intrathecal application of a miR-1306-3p antagomir and A317491 significantly alleviated visceral pain in a rat model of chronic visceral pain. Together, our findings suggest that miR-1306-3p might function as an endogenous ligand to activate P2X3Rs, eventually leading to chronic visceral pain.
Collapse
Affiliation(s)
- Yan-Yan Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, P.R. China
- School of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, P. R. China
| | - Qian Wang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, P.R. China
| | - Ping-An Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Cheng Zhu
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, P.R. China
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
44
|
Xiong J, Fu F, Yu F, He X. Advances of exosomal miRNAs in the diagnosis and treatment of ovarian cancer. Discov Oncol 2023; 14:65. [PMID: 37160813 PMCID: PMC10169985 DOI: 10.1007/s12672-023-00674-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 04/27/2023] [Indexed: 05/11/2023] Open
Abstract
Ovarian cancer is a tumor with the highest fatalities among female malignant tumors. This disease has no typical symptoms in its early stage, and most of the patients are in an advanced stage when being treated. The treatment effect is poor and it is easy to develop chemotherapy resistance. Therefore, it is particularly urgent to clarify the pathogenesis of ovarian cancer, explore its early diagnosis of biomarkers, and discover new treatment methods. As a carrier of intercellular information and genetic material transfer, exosomes are widely distributed in body fluids (e.g. blood and urine), which are regarded as latent tumor markers and take effects on tumor occurrence and invasion. Several articles have recently signified that exosomal miRNAs are widely implicated in the formation of the ovarian cancer tumor microenvironment, disease initiation and progression, and the generation of chemotherapy resistance. This article reviews the research on exosomal miRNAs in ovarian cancer.
Collapse
Affiliation(s)
- Jun Xiong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, NanChang, JiangXi, China
| | - Fen Fu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, NanChang, JiangXi, China
| | - Feng Yu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, NanChang, JiangXi, China
| | - Xiaoju He
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, NanChang, JiangXi, China.
| |
Collapse
|
45
|
Christodoulou MI, Zaravinos A. Single-Cell Analysis in Immuno-Oncology. Int J Mol Sci 2023; 24:8422. [PMID: 37176128 PMCID: PMC10178969 DOI: 10.3390/ijms24098422] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
The complexity of the cellular and non-cellular milieu surrounding human tumors plays a decisive role in the course and outcome of disease. The high variability in the distribution of the immune and non-immune compartments within the tumor microenvironments (TME) among different patients governs the mode of their response or resistance to current immunotherapeutic approaches. Through deciphering this diversity, one can tailor patients' management to meet an individual's needs. Single-cell (sc) omics technologies have given a great boost towards this direction. This review gathers recent data about how multi-omics profiling, including the utilization of single-cell RNA sequencing (scRNA-seq), assay for transposase-accessible chromatin with sequencing (scATAC-seq), T-cell receptor sequencing (scTCR-seq), mass, tissue-based, or microfluidics cytometry, and related bioinformatics tools, contributes to the high-throughput assessment of a large number of analytes at single-cell resolution. Unravelling the exact TCR clonotype of the infiltrating T cells or pinpointing the classical or novel immune checkpoints across various cell subsets of the TME provide a boost to our comprehension of adaptive immune responses, their antigen specificity and dynamics, and grant suggestions for possible therapeutic targets. Future steps are expected to merge high-dimensional data with tissue localization data, which can serve the investigation of novel multi-modal biomarkers for the selection and/or monitoring of the optimal treatment from the current anti-cancer immunotherapeutic armamentarium.
Collapse
Affiliation(s)
- Maria-Ioanna Christodoulou
- Tumor Immunology and Biomarkers Group, Basic and Translational Cancer Research Center (BTCRC), 1516 Nicosia, Cyprus
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus
- Cancer Genetics, Genomics and Systems Biology Group, Basic and Translational Cancer Research Center (BTCRC), 1516 Nicosia, Cyprus
| |
Collapse
|
46
|
Song D, Wang F, Ju Y, He Q, Sun T, Deng W, Ding R, Zhang C, Xu Q, Qi C, Bao J. Application and development of noninvasive biomarkers for colorectal cancer screening: a systematic review. Int J Surg 2023; 109:925-935. [PMID: 36974713 PMCID: PMC10389553 DOI: 10.1097/js9.0000000000000260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/22/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is the second most common cause of cancer-related death (9.4% of the 9.9 million cancer deaths). However, CRC develops slowly, and early detection and intervention can effectively improve the survival rate and quality of life. Although colonoscopy can detect and diagnose CRC, it is unsuitable for CRC screening in average-risk populations. Some commercial kits based on DNA mutation or methylation are approved for screening, but the low sensitivity for advanced adenoma or early-stage CRC would limit the applications. MAIN RESULTS Recently, researchers have focused on developing noninvasive or minimally invasive, easily accessible biomarkers with higher sensitivity and accuracy for CRC screening. Numerous reports describe advances in biomarkers, including DNA mutations and methylation, mRNA and miRNA, gut microbes, and metabolites, as well as low-throughput multiomics panels. In small cohorts, the specificity and sensitivity improved when fecal immunochemical testing combined with other biomarkers; further verification in large cohorts is expected. In addition, the continuous improvement of laboratory technology has also improved the sensitivity of detection technology, such as PCR, and the application of CRISPR/Cas technology. Besides, artificial intelligence has extensively promoted the mining of biomarkers. Machine learning was performed to construct a diagnosis model for CRC screening based on the cfDNA fragment features from whole-genome sequencing data. In another study, multiomics markers, including cfDNA, epigenetic, and protein signals, were also discovered by machine learning. Finally, advancements in sensor technology promote the applicability of volatile organic compounds in CRC early detection. CONCLUSION Here, the authors review advances in early detection and screening of CRC based on different biomarker types. Most studies reported optimistic findings based on preliminary research, and prospective clinical studies are ongoing. These promising biomarkers are expected to more accurately identify early-stage patients with CRC and be applied in the future.
Collapse
Affiliation(s)
| | - Fei Wang
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd, Nanjing Simcere Medical Laboratory Science Co. Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd, Xuanwu District, Nanjing, Jiangsu Province, China
| | - Yongzhi Ju
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd, Nanjing Simcere Medical Laboratory Science Co. Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd, Xuanwu District, Nanjing, Jiangsu Province, China
| | - Qianru He
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd, Nanjing Simcere Medical Laboratory Science Co. Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd, Xuanwu District, Nanjing, Jiangsu Province, China
| | - Tingting Sun
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd, Nanjing Simcere Medical Laboratory Science Co. Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd, Xuanwu District, Nanjing, Jiangsu Province, China
| | - Wanglong Deng
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd, Nanjing Simcere Medical Laboratory Science Co. Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd, Xuanwu District, Nanjing, Jiangsu Province, China
| | - Ran Ding
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd, Nanjing Simcere Medical Laboratory Science Co. Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd, Xuanwu District, Nanjing, Jiangsu Province, China
| | - Chao Zhang
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd, Nanjing Simcere Medical Laboratory Science Co. Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd, Xuanwu District, Nanjing, Jiangsu Province, China
| | - Qing Xu
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd, Nanjing Simcere Medical Laboratory Science Co. Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd, Xuanwu District, Nanjing, Jiangsu Province, China
| | - Chuang Qi
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd, Nanjing Simcere Medical Laboratory Science Co. Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd, Xuanwu District, Nanjing, Jiangsu Province, China
| | - Jun Bao
- Medical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting
| |
Collapse
|
47
|
Zhang H, Liu L, Liu J, Dang P, Hu S, Yuan W, Sun Z, Liu Y, Wang C. Roles of tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for solid cancers. Mol Cancer 2023; 22:58. [PMID: 36941614 PMCID: PMC10029244 DOI: 10.1186/s12943-023-01725-x] [Citation(s) in RCA: 142] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/16/2023] [Indexed: 03/23/2023] Open
Abstract
In recent years, tumor immunotherapy has made significant progress. However, tumor immunotherapy, particularly immune checkpoint inhibitors (e.g., PD-1/PD-L1 inhibitors), benefits only a tiny proportion of patients in solid cancers. The tumor microenvironment (TME) acts a significant role in tumor immunotherapy. Studies reported that tumor-associated macrophages (TAMs), as one of the main components of TME, seriously affected the therapeutic effect of PD-1/PD-L1 inhibitors. In this review, we analyzed TAMs from epigenetic and single-cell perspectives and introduced the role and mechanisms of TAMs in anti-programmed death protein 1(anti-PD-1) therapy. In addition, we summarized combination regimens that enhance the efficacy of tumor PD-1/PD-L1 inhibitors and elaborated on the role of the TAMs in different solid cancers. Eventually, the clinical value of TAMs by influencing the therapeutic effect of tumor PD-1/PD-L1 inhibitors was discussed. These above are beneficial to elucidate poor therapeutic effect of PD-1/PD-L1 inhibitors in solid tumors from the point of view of TAMs and explore the strategies to improve its objective remission rate of solid cancers.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Lin Liu
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jinbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Pengyuan Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China.
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yang Liu
- Department of Radiotherapy, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450001, China.
| | - Chengzeng Wang
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
48
|
Missiaen R, Lesner NP, Simon MC. HIF: a master regulator of nutrient availability and metabolic cross-talk in the tumor microenvironment. EMBO J 2023; 42:e112067. [PMID: 36808622 PMCID: PMC10015374 DOI: 10.15252/embj.2022112067] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 02/22/2023] Open
Abstract
A role for hypoxia-inducible factors (HIFs) in hypoxia-dependent regulation of tumor cell metabolism has been thoroughly investigated and covered in reviews. However, there is limited information available regarding HIF-dependent regulation of nutrient fates in tumor and stromal cells. Tumor and stromal cells may generate nutrients necessary for function (metabolic symbiosis) or deplete nutrients resulting in possible competition between tumor cells and immune cells, a result of altered nutrient fates. HIF and nutrients in the tumor microenvironment (TME) affect stromal and immune cell metabolism in addition to intrinsic tumor cell metabolism. HIF-dependent metabolic regulation will inevitably result in the accumulation or depletion of essential metabolites in the TME. In response, various cell types in the TME will respond to these hypoxia-dependent alterations by activating HIF-dependent transcription to alter nutrient import, export, and utilization. In recent years, the concept of metabolic competition has been proposed for critical substrates, including glucose, lactate, glutamine, arginine, and tryptophan. In this review, we discuss how HIF-mediated mechanisms control nutrient sensing and availability in the TME, the competition for nutrients, and the metabolic cross-talk between tumor and stromal cells.
Collapse
Affiliation(s)
- Rindert Missiaen
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas P Lesner
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
49
|
Yang C, Li D, Ko CN, Wang K, Wang H. Active ingredients of traditional Chinese medicine for enhancing the effect of tumor immunotherapy. Front Immunol 2023; 14:1133050. [PMID: 36969211 PMCID: PMC10036358 DOI: 10.3389/fimmu.2023.1133050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Immunotherapy is a type of treatment that uses our own immune system to fight cancer. Studies have shown that traditional Chinese medicine (TCM) has antitumor activity and can enhance host immunity. This article briefly describes the immunomodulatory and escape mechanisms in tumors, as well as highlights and summarizes the antitumor immunomodulatory activities of some representative active ingredients of TCM. Finally, this article puts forward some opinions on the future research and clinical application of TCM, aiming to promote the clinical applications of TCM in tumor immunotherapy and to provide new ideas for the research of tumor immunotherapy using TCM.
Collapse
Affiliation(s)
- Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, Zhejiang, China
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chung-Nga Ko
- C-MER Dennis Lam and Partners Eye Center, Hong Kong International Eye Care Group, Hong Kong, China
- *Correspondence: Chung-Nga Ko, ; Kai Wang, ; Haiyong Wang,
| | - Kai Wang
- Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
- *Correspondence: Chung-Nga Ko, ; Kai Wang, ; Haiyong Wang,
| | - Haiyong Wang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Chung-Nga Ko, ; Kai Wang, ; Haiyong Wang,
| |
Collapse
|
50
|
Fan Y, Liao J, Wang Y, Wang Z, Zheng H, Wang Y. miR-132-3p regulates antibody-mediated complement-dependent cytotoxicity in colon cancer cells by directly targeting CD55. Clin Exp Immunol 2023; 211:57-67. [PMID: 36571232 PMCID: PMC9993456 DOI: 10.1093/cei/uxac120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/18/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022] Open
Abstract
The overexpression of membrane-bound complement regulatory proteins (mCRPs) on tumour cells helps them survive complement attacks by suppressing antibody-mediated complement-dependent cytotoxicity (CDC). Consequently, mCRP overexpression limits monoclonal antibody drug immune efficacy. CD55, an mCRP, plays an important role in inhibiting antibody-mediated CDC. However, the mechanisms regulating CD55 expression in tumour cells remain unclear. Here, the aim was to explore CD55-targeting miRNAs. We previously constructed an in vitro model comprising cancer cell lines expressing α-gal and serum containing natural antibodies against α-gal and complement. This was used to simulate antibody-mediated CDC in colon cancer cells. We screened microRNAs that directly target CD55 using LoVo and Ls-174T colon cell lines, which express CD55 at low and high levels, respectively. miR-132-3p expression was dramatically lower in Ls-174T cells than in LoVo cells. miR-132-3p overexpression or inhibition transcriptionally regulated CD55 expression by specifically targeting its mRNA 3'-untranslated regions. Further, miR-132-3p modulation regulated colon cancer cell sensitivity to antibody-mediated CDC through C5a release and C5b-9 deposition. Moreover, miR-132-3p expression was significantly reduced, whereas CD55 expression was increased, in colon cancer tissues compared to levels in adjacent normal tissues. CD55 protein levels were negatively correlated with miR-132-3p expression in colon cancer tissues. Our results indicate that miR-132-3p regulates colon cancer cell sensitivity to antibody-mediated CDC by directly targeting CD55. In addition, incubating the LoVo human tumour cell line, stably transfected with the xenoantigen α-gal, with human serum containing natural antibodies comprises a stable and cheap in vitro model to explore the mechanisms underlying antibody-mediated CDC.
Collapse
Affiliation(s)
- Yu Fan
- Multi-omics Laboratory of Breast Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Juan Liao
- Multi-omics Laboratory of Breast Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Wang
- Multi-omics Laboratory of Breast Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhu Wang
- Multi-omics Laboratory of Breast Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zheng
- Multi-omics Laboratory of Breast Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanping Wang
- Correspondence: Yanping Wang, 5# Gongxing Street, Chengdu, Sichuan, China.
| |
Collapse
|