1
|
O'Gorman B, Cooke SF, Blair CM. Precision peptide disruptors: The next generation of targeted therapeutics in oncology. Cell Signal 2025; 132:111783. [PMID: 40180165 DOI: 10.1016/j.cellsig.2025.111783] [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: 01/07/2025] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Therapeutically targeting the pathologically remodelled protein-protein interaction network in cancer with peptide disruptors increasingly represents a clinically attractive approach to treating recalcitrant cancers. In this review, we map the pre-clinical and clinical-stage peptide disruptor landscape within an oncology-specific context and discuss key clinical examples that are making significant impact to patients; demonstrating a key role for peptide disruptors in precision medicine as a next-generation targeted therapeutic.
Collapse
Affiliation(s)
- Bria O'Gorman
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Sean F Cooke
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Connor M Blair
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
2
|
Kwan A, Mcdermott-Brown I, Muthana M. Proliferating Cell Nuclear Antigen in the Era of Oncolytic Virotherapy. Viruses 2024; 16:1264. [PMID: 39205238 PMCID: PMC11359830 DOI: 10.3390/v16081264] [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: 06/10/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Proliferating cell nuclear antigen (PCNA) is a well-documented accessory protein of DNA repair and replication. It belongs to the sliding clamp family of proteins that encircle DNA and acts as a mobile docking platform for interacting proteins to mount and perform their metabolic tasks. PCNA presence is ubiquitous to all cells, and when located in the nucleus it plays a role in DNA replication and repair, cell cycle control and apoptosis in proliferating cells. It also plays a crucial role in the infectivity of some viruses, such as herpes simplex viruses (HSVs). However, more recently it has been found in the cytoplasm of immune cells such as neutrophils and macrophages where it has been shown to be involved in the development of a pro-inflammatory state. PCNA is also expressed on the surface of certain cancer cells and can play a role in preventing immune cells from killing tumours, as well as being associated with cancer virulence. Given the growing interest in oncolytic viruses (OVs) as a novel cancer therapeutic, this review considers the role of PCNA in healthy, cancerous, and immune cells to gain an understanding of how PCNA targeted therapy and oncolytic virotherapy may interact in the future.
Collapse
Affiliation(s)
| | | | - Munitta Muthana
- Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (A.K.)
| |
Collapse
|
3
|
Søgaard CK, Otterlei M. Targeting proliferating cell nuclear antigen (PCNA) for cancer therapy. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:209-246. [PMID: 39034053 DOI: 10.1016/bs.apha.2024.04.002] [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: 07/23/2024]
Abstract
Proliferating cell nuclear antigen (PCNA) is an essential scaffold protein in many cellular processes. It is best known for its role as a DNA sliding clamp and processivity factor during DNA replication, which has been extensively reviewed by others. However, the importance of PCNA extends beyond its DNA-associated functions in DNA replication, chromatin remodelling, DNA repair and DNA damage tolerance (DDT), as new non-canonical roles of PCNA in the cytosol have recently been identified. These include roles in the regulation of immune evasion, apoptosis, metabolism, and cellular signalling. The diverse roles of PCNA are largely mediated by its myriad protein interactions, and its centrality to cellular processes makes PCNA a valid therapeutic anticancer target. PCNA is expressed in all cells and plays an essential role in normal cellular homeostasis; therefore, the main challenge in targeting PCNA is to selectively kill cancer cells while avoiding unacceptable toxicity to healthy cells. This chapter focuses on the stress-related roles of PCNA, and how targeting these PCNA roles can be exploited in cancer therapy.
Collapse
Affiliation(s)
- Caroline K Søgaard
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Otterlei
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway; APIM Therapeutics A/S, Trondheim, Norway.
| |
Collapse
|
4
|
Tan Y, Ding L, Li G. MCM4 acts as a biomarker for LUAD prognosis. J Cell Mol Med 2023; 27:3354-3362. [PMID: 37817427 PMCID: PMC10623528 DOI: 10.1111/jcmm.17819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 04/16/2023] [Accepted: 06/14/2023] [Indexed: 10/12/2023] Open
Abstract
MCM4 forms the pre-replication complex (MCM2-7) with five other minichromosome maintenance (MCM) proteins. This complex binds to replication origins at G1 stage in cell cycle process, playing a critical role in DNA replication initiation. Recently, MCM4 is reported to have a complex interaction with multiple cancer progression, including gastric, ovarian and cervical cancer. Here, this study mainly focused on the expression of MCM4 and its values in lung adenocarcinoma (LUAD). MCM4 was highly expressed in LUAD tumours and cells, and had an important effect on the overall survival. Overexpression of MCM4 promoted the proliferation, and suppressed the apoptosis in LUAD cells. However, MCM4 silence led to the opposite results. In vivo, knockdown of MCM4 inhibited tumour volume and weight in xenograft mouse model. As a member of DNA helicase, knockdown of MCM4 caused cell cycle arrest at G1 stage through inducing the expression of P21, a CDK inhibitor. These findings indicate that MCM4 may be a possible new therapeutic target for LUAD in the future.
Collapse
Affiliation(s)
- Yue Tan
- Branch of Minhang, Department of Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Lei Ding
- Department of Ultrasonic DiagnosisSecond Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Guiyuan Li
- Department of Oncology, School of Medicine, Tongji HospitalTongji UniversityShanghaiChina
| |
Collapse
|
5
|
NKp44-Derived Peptide Used in Combination Stimulates Antineoplastic Efficacy of Targeted Therapeutic Drugs. Int J Mol Sci 2022; 23:ijms232214054. [PMID: 36430528 PMCID: PMC9692391 DOI: 10.3390/ijms232214054] [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: 10/28/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Lung cancer cells in the tumor microenvironment facilitate immune evasion that leads to failure of conventional chemotherapies, despite provisionally decided on the genetic diagnosis of patients in a clinical setup. The current study follows three lung cancer patients who underwent "personalized" chemotherapeutic intervention. Patient-derived xenografts (PDXs) were subjected to tumor microarray and treatment screening with chemotherapies, either individually or in combination with the peptide R11-NLS-pep8; this peptide targets both membrane-associated and nuclear PCNA. Ex vivo, employing PDX-derived explants, it was found that combination with R11-NLS-pep8 stimulated antineoplastic effect of chemotherapies that were, although predicted based on the patient's genetic mutation, inactive on their own. Furthermore, treatment in vivo of PDX-bearing mice showed an exactly similar trend in the result, corroborating the finding to be translated into clinical setup.
Collapse
|
6
|
Sosa Cuevas E, Valladeau-Guilemond J, Mouret S, Roubinet B, de Fraipont F, Landemarre L, Charles J, Bendriss-Vermare N, Chaperot L, Aspord C. Unique CLR expression patterns on circulating and tumor-infiltrating DC subsets correlated with clinical outcome in melanoma patients. Front Immunol 2022; 13:1040600. [PMID: 36353633 PMCID: PMC9638162 DOI: 10.3389/fimmu.2022.1040600] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/10/2022] [Indexed: 08/15/2023] Open
Abstract
Subversion of immunity by tumors is a crucial step for their development. Dendritic cells (DCs) are strategic immune cells that orchestrate anti-tumor immune responses but display altered functions in cancer. The bases for such DCs' hijacking are not fully understood. Tumor cells harbor unusual glycosylation patterns of surface glycoproteins and glycolipids. DCs express glycan-binding receptors, named C-type lectin receptors (CLR), allowing them to sense changes in glycan signature of their environment, and subsequently trigger a response. Recognition of tumor glycans by CLRs is crucial for DCs to shape antitumor immunity, and decisive in the orientation of the response. Yet the status of the CLR machinery on DCs in cancer, especially melanoma, remained largely unknown. We explored CLR expression patterns on circulating and tumor-infiltrating cDC1s, cDC2s, and pDCs of melanoma patients, assessed their clinical relevance, and further depicted the correlations between CLR expression profiles and DCs' features. For the first time, we highlighted that the CLR repertoire of circulating and tumor-infiltrating cDC1s, cDC2s, and pDCs was strongly perturbed in melanoma patients, with modulation of DCIR, CLEC-12α and NKp44 on circulating DCs, and perturbation of Dectin-1, CD206, DEC205, DC-SIGN and CLEC-9α on tumor-infiltrating DCs. Furthermore, melanoma tumor cells directly altered CLR expression profiles of healthy DC subsets, and this was associated with specific glycan patterns (Man, Fuc, GlcNAc) that may interact with DCs through CLR molecules. Notably, specific CLR expression profiles on DC subsets correlated with unique DCs' activation status and functionality and were associated with clinical outcome of melanoma patients. Higher proportions of DCIR-, DEC205-, CLEC-12α-expressing cDCs were linked with a better survival, whereas elevated proportions of CD206-, Dectin1-expressing cDCs and NKp44-expressing pDCs were associated with a poor outcome. Thus, melanoma tumor may shape DCs' features by exploiting the plasticity of the CLR machinery. Our study revealed that melanoma manipulates CLR pathways to hijack DC subsets and escape from immune control. It further paved the way to exploit glycan-lectin interactions for the design of innovative therapeutic strategies, which exploit DCs' potentialities while avoiding hijacking by tumor, to properly reshape anti-tumor immunity by manipulating the CLR machinery.
Collapse
Affiliation(s)
- Eleonora Sosa Cuevas
- Etablissement Français du Sang Auvergne-Rhône-Alpes, R&D Laboratory, Grenoble, France
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Jenny Valladeau-Guilemond
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Stephane Mouret
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Grenoble, France
| | | | - Florence de Fraipont
- Medical Unit of Molecular genetic (Hereditary Diseases and Oncology), Grenoble University Hospital, Grenoble, France
| | | | - Julie Charles
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Grenoble, France
| | - Nathalie Bendriss-Vermare
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Laurence Chaperot
- Etablissement Français du Sang Auvergne-Rhône-Alpes, R&D Laboratory, Grenoble, France
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Caroline Aspord
- Etablissement Français du Sang Auvergne-Rhône-Alpes, R&D Laboratory, Grenoble, France
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
7
|
Cai D, Ma X, Guo H, Zhang H, Bian A, Yu H, Cheng W. Prognostic value of p16, p53, and pcna in sarcoma and an evaluation of immune infiltration. J Orthop Surg Res 2022; 17:305. [PMID: 35689249 PMCID: PMC9185979 DOI: 10.1186/s13018-022-03193-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/26/2022] [Indexed: 12/22/2022] Open
Abstract
Background p16, p53, and proliferating cell nuclear antigen (pcna) genes play significant roles in many chromatin modifications and have been found to be highly expressed in a variety of tumor tissues. Therefore, they have been used as target genes for some tumor therapies. However, the differential expressions of the p16, p53, and pcna genes in human sarcomas and their effects on prognosis have not been widely reported. Methods The Oncomine dataset was used to analyze the transcription levels of p16, p53, and pcna genes, and the gene expression profile interactive analysis (GEPIA) dataset was used to analyze the differential expressions of p16, p53, and pcna. The expression levels of p16, p53, and pcna were further analyzed by Western Blotting. GEPIA and Kaplan–Meier analyses were used to analyze the prognostic value of p16, p53, and pcna. Furthermore, p16, p53, and pcna gene mutations and their association with overall survival (OS) and disease-free survival (DFS) were analyzed using cBioPortal datasets. In addition, genes co-expressed with p16, p53, and pcna were analyzed using Oncomine. The DAVID dataset was used to analyze the functional enrichment of p16, p53, pcna, and their co-expressed genes by Gene Ontology (GO) and Metascape were used to construct a network map. Finally, the immune cell infiltration of p16, p53, and pcna in patients with sarcoma was reported by Tumor Immune Estimation Resource (TIMER). Results p16, p53, and pcna were up-regulated in human sarcoma tissues and almost all sarcoma cell lines. Western Blotting showed that the expression of p16, p53, and pcna was elevated in osteosarcoma cell lines. The expression of pcna was correlated with OS, the expression of p16, p53, and pcna was correlated with relapse-free survival, and the genetic mutation of p16 was negatively correlated with OS and DFS. We also found that p16, p53, and pcna genes were positively/negatively correlated with immune cell infiltration in sarcoma. Conclusions The results of this study showed that p16, p53, and pcna can significantly affect the survival and immune status of sarcoma patients. Therefore, p16, p53, and pcna could be used as potential biomarkers of prognosis and immune infiltration in human sarcoma and provide a possible therapeutic target for sarcoma.
Collapse
Affiliation(s)
- Dechao Cai
- Department of Orthopedics, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, China
| | - Xiao Ma
- Department of Orthopedics, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, China
| | - Huihui Guo
- Department of Orthopedics, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, China
| | - Haotian Zhang
- Department of Orthopedics, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, China
| | - Ashuai Bian
- Department of Orthopedics, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, China
| | - Haoran Yu
- Department of Orthopedics, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, China
| | - Wendan Cheng
- Department of Orthopedics, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, China.
| |
Collapse
|
8
|
Zorko M, Jones S, Langel Ü. Cell-penetrating peptides in protein mimicry and cancer therapeutics. Adv Drug Deliv Rev 2022; 180:114044. [PMID: 34774552 DOI: 10.1016/j.addr.2021.114044] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 12/14/2022]
Abstract
Extensive research has been undertaken in the pursuit of anticancer therapeutics. Many anticancer drugs require specificity of delivery to cancer cells, whilst sparing healthy tissue. Cell-penetrating peptides (CPPs), now well established as facilitators of intracellular delivery, have in recent years advanced to incorporate target specificity and thus possess great potential for the targeted delivery of anticancer cargoes. Though none have yet been approved for clinical use, this novel technology has already entered clinical trials. In this review we present CPPs, discuss their classification, mechanisms of cargo internalization and highlight strategies for conjugation to anticancer moieties including their incorporation into therapeutic proteins. As the mainstay of this review, strategies to build specificity into tumor targeting CPP constructs through exploitation of the tumor microenvironment and the use of tumor homing peptides are discussed, whilst acknowledging the extensive contribution made by CPP constructs to target specific protein-protein interactions integral to intracellular signaling pathways associated with tumor cell survival and progression. Finally, antibody/antigen CPP conjugates and their potential roles in cancer immunotherapy and diagnostics are considered. In summary, this review aims to harness the potential of CPP-aided drug delivery for future cancer therapies and diagnostics whilst highlighting some of the most recent achievements in selective delivery of anticancer drugs, including cytostatic drugs, to a range of tumor cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Matjaž Zorko
- University of Ljubljana, Medical Faculty, Institute of Biochemistry and Molecular Genetics, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Sarah Jones
- University of Wolverhampton, School of Pharmacy, Faculty of Science & Engineering, Wulfruna Street, Wolverhampton WV1 1LY, UK.
| | - Ülo Langel
- University of Stockholm, Department of Biochemistry and Biophysics, Svante Arrhenius väg 16, 106 91 Stockholm, Sweden; Institute of Technology, University of Tartu, Nooruse 1, Tartu, Estonia 50411, Estonia.
| |
Collapse
|
9
|
Ammar OA, El-Missiry MA, Othman AI, Amer ME. Melatonin is a potential oncostatic agent to inhibit HepG2 cell proliferation through multiple pathways. Heliyon 2022; 8:e08837. [PMID: 35141433 PMCID: PMC8814902 DOI: 10.1016/j.heliyon.2022.e08837] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/22/2021] [Accepted: 01/24/2022] [Indexed: 11/25/2022] Open
Abstract
CONTEXT Chemotherapy is a cornerstone in the treatment of hepatocellular carcinoma (HCC). Melatonin is a pineal hormone that targets various cancers, however, its antitumor pathways are still not fully elucidated. OBJECTIVE This study investigated melatonin's antitumor molecular mechanisms to inhibit the proliferation of HepG2 cells. MATERIALS AND METHODS HepG2 Cells were classified into cells without treatment as a control group and cells treated with melatonin (5.4 mmol/L) for 48 h. Proliferating cell nuclear antigen (PCNA) and marker of proliferation Ki-67 were estimated using immunohistochemical analysis. Apoptosis and cell cycle were evaluated using flow cytometric analysis. Apoptotic markers were detected using RT-qPCR assay. Antioxidants and oxidative stress biomarkers were performed using a colorimetric assay. RESULTS Melatonin produced a remarkable steady decrease in the viability of HepG2 cells at a concentration range between 5-20 mmol/L. Melatonin suppressed cell proliferation in the G2/M phase of the cell cycle (34.97 ± 0.92%) and induced apoptosis (12.43 ± 0.73%) through up-regulating p21 and p53 that was confirmed by the reduction of PCNA and Ki-67 expressions. Additionally, melatonin repressed angiogenesis evidenced by the down-regulation of angiopoietin-2, vascular endothelial growth factor receptor-2 expressions (0.42-fold change), and the level of CD133. Moreover, melatonin augmented the oxidative stress manifested by a marked increase of 4-hydroxynonenal levels with a reduction of glutathione content and superoxide dismutase activity. DISCUSSION AND CONCLUSION Melatonin inhibits proliferation and angiogenesis and induced apoptosis and oxidative stress in HepG2 cells. These results indicate the oncostatic effectiveness of melatonin on liver cancer.
Collapse
Affiliation(s)
- Omar A. Ammar
- Basic Science Department, Delta University for Science and Technology, Gamasa, Egypt
| | | | - Azza I. Othman
- Zoology Department, Faculty of Science, Mansoura University, Egypt
| | - Maggie E. Amer
- Zoology Department, Faculty of Science, Mansoura University, Egypt
| |
Collapse
|
10
|
Girard P, Sosa Cuevas E, Ponsard B, Mouret S, Gil H, Col E, De Fraipont F, Sturm N, Charles J, Manches O, Chaperot L, Aspord C. Dysfunctional BTN3A together with deregulated immune checkpoints and type I/II IFN dictate defective interplay between pDCs and γδ T cells in melanoma patients, which impacts clinical outcomes. Clin Transl Immunology 2021; 10:e1329. [PMID: 34786191 PMCID: PMC8577077 DOI: 10.1002/cti2.1329] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/22/2021] [Accepted: 07/29/2021] [Indexed: 01/01/2023] Open
Abstract
Objectives pDCs and γδ T cells emerge as potent immune players participating in the pathophysiology of cancers, yet still remaining enigmatic while harbouring a promising potential for clinical translations. Despite strategic and closed missions, crosstalk between pDCs and γδ T cells has not been deciphered yet in cancers, especially in melanoma where the long‐term control of the tumor still remains a challenge. Methods This prompted us to explore the interplay between pDCs and γδ T cells in the context of melanoma, investigating the reciprocal features of pDCs or γδ T cells, the underlying molecular mechanisms and its impact on clinical outcomes. Results TLRL‐activated pDCs from the blood and tumor infiltrate of melanoma patients displayed an impaired ability to activate, to modulate immune checkpoints and trigger the functionality of γδ T cells. Conversely, γδ T cells from the blood or tumor infiltrate of melanoma patients activated by PAg were defective in triggering pDCs’ activation and modulation of immune checkpoints, and failed to elicit the functionality of pDCs. Reversion of the dysfunctional cross‐talks could be achieved by specific cytokine administration and immune checkpoint targeting. Strikingly, we revealed an increased expression of BTN3A on circulating and tumor‐infiltrating pDCs and γδ T cells from melanoma patients, but stressed out the potential impairment of this molecule. Conclusion Our study uncovered that melanoma hijacked the bidirectional interplay between pDCs and γδ T cells to escape from immune control, and revealed BTN3A dysfunction. Such understanding will help harness and synergise the power of these potent immune cells to design new therapeutic approaches exploiting their antitumor potential while counteracting their skewing by tumors to improve patient outcomes.
Collapse
Affiliation(s)
- Pauline Girard
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,Etablissement Français du Sang Auvergne-Rhône-Alpes R&D Laboratory Grenoble France
| | - Eleonora Sosa Cuevas
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,Etablissement Français du Sang Auvergne-Rhône-Alpes R&D Laboratory Grenoble France
| | - Benedicte Ponsard
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,Etablissement Français du Sang Auvergne-Rhône-Alpes R&D Laboratory Grenoble France
| | - Stephane Mouret
- Dermatology Clinic Grenoble University Hospital Grenoble France
| | - Hugo Gil
- Pathology Department Institut de Biologie et Pathologie CHU Grenoble Alpes Grenoble France
| | - Edwige Col
- Pathology Department Institut de Biologie et Pathologie CHU Grenoble Alpes Grenoble France
| | - Florence De Fraipont
- Medical Unit of Molecular Genetic (Hereditary Diseases and Oncology) Grenoble University Hospital Grenoble France
| | - Nathalie Sturm
- Pathology Department Institut de Biologie et Pathologie CHU Grenoble Alpes Grenoble France
| | - Julie Charles
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,Dermatology Clinic Grenoble University Hospital Grenoble France
| | - Olivier Manches
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,Etablissement Français du Sang Auvergne-Rhône-Alpes R&D Laboratory Grenoble France
| | - Laurence Chaperot
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,Etablissement Français du Sang Auvergne-Rhône-Alpes R&D Laboratory Grenoble France
| | - Caroline Aspord
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,Etablissement Français du Sang Auvergne-Rhône-Alpes R&D Laboratory Grenoble France
| |
Collapse
|
11
|
Exosomal microRNA-15a from mesenchymal stem cells impedes hepatocellular carcinoma progression via downregulation of SALL4. Cell Death Discov 2021; 7:224. [PMID: 34455417 PMCID: PMC8403170 DOI: 10.1038/s41420-021-00611-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 06/21/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a heterogeneous tumor with an increased incidence worldwide accompanied by high mortality and dismal prognosis. Emerging evidence indicates that mesenchymal stem cells (MSCs)-derived exosomes possess protective effects against various human diseases by transporting microRNAs (miRNAs or miRs). We aimed to explore the role of exosomal miR-15a derived from MSCs and its related mechanisms in HCC. Exosomes were isolated from transduced MSCs and co-incubated with Hep3B and Huh7 cells. miR-15a expression was examined by RT-qPCR in HCC cells, MSCs, and secreted exosomes. CCK-8, transwell, and flow cytometry were used to detect the effects of miR-15a or spalt-like transcription factor 4 (SALL4) on cell proliferative, migrating, invasive, and apoptotic properties. A dual-luciferase reporter gene assay was performed to validate the predicted targeting relationship of miR-15a with SALL4. Finally, in vivo experiments in nude mice were implemented to assess the impact of exosome-delivered miR-15a on HCC. The exosomes from MSCs restrained HCC cell proliferative, migrating, and invasive potentials, and accelerated their apoptosis. miR-15a was expressed at low levels in HCC cells and could bind to SALL4, thus curtailing the proliferative, migrating, and invasive abilities of HCC cells. Exosomes successfully delivered miR-15a to HCC cells. Exosomal miR-15a depressed tumorigenicity and metastasis of HCC tumors in vivo. Overall, exosomal miR-15a from MSCs can downregulate SALL4 expression and thereby retard HCC development.
Collapse
|
12
|
Li P, Wang L, Li P, Hu F, Cao Y, Tang D, Ye G, Li H, Wang D. Silencing of long non-coding RNA XIST represses gastric cancer progression through blocking NFκB pathway via inhibiting HNF4A-mediated transcription of EPHA1. Cancer Gene Ther 2021; 28:307-320. [PMID: 33199830 DOI: 10.1038/s41417-020-00220-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/05/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
Gastric cancer (GC) is a common cancer and a leading cause of cancer-related deaths worldwide. Recent studies have supported the important role of long non-coding RNAs (lncRNAs) in GC progression. This study identified functional significance of X inactive specific transcript (XIST) in GC. The expression of XIST and EPHA1 in GC tissues and cells was measured. Then, dual luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay and Chromatin Immunoprecipitation (ChIP) assay were performed to explore the interaction among XIST, EPHA1 and HNF4A. The effects of XIST on GG progression were evaluated by determining expression of proliferation- and invasion-related proteins (Ki67, PCNA, MMP-2, and MMP-9). Further, the functional role of XIST in GC with the involvement of NFκB pathway was also analyzed. Subsequently, the tumor growth in nude mice was evaluated. High expression of XIST and EPHA1 was observed in GC. XIST elevated EPHA1 expression by recruiting HNF4A. In addition, silencing of XIST inhibited GC progression in vitro and in vivo. Overexpressed XIST and EPHA1 yielded a reversed effect on cell proliferation and invasion. SN50 treatment (inhibitor of NFκB pathway) counteracted the promotive effect on GC cell proliferation and invasion mediated by XIST. The present study unveils that XIST increases the enrichment of HNF4A in the promoter region of EPHA1, thus promoting the deterioration of GC.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Proliferation/physiology
- Disease Progression
- Hepatocyte Nuclear Factor 4/antagonists & inhibitors
- Hepatocyte Nuclear Factor 4/metabolism
- Heterografts
- Humans
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NF-kappa B/metabolism
- RNA, Long Noncoding/antagonists & inhibitors
- RNA, Long Noncoding/biosynthesis
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Receptor, EphA1/genetics
- Receptor, EphA1/metabolism
- Signal Transduction
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Transcription, Genetic
Collapse
Affiliation(s)
- Ping Li
- Department of Central Laboratory, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
- Department of General Surgery, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
- Department of Experimental Surgery-Cancer Metastasis, Medical Faculty Mannheim, Ruprecht Karls University, Mannheim, 68167, Germany
| | - Liuhua Wang
- Department of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Institute of General Surgery-Yangzhou, Yangzhou University, Yangzhou, 225000, PR China
| | - Pengfei Li
- Department of Central Laboratory, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
- Department of General Surgery, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
| | - Fangyong Hu
- Department of Central Laboratory, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
- Department of General Surgery, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
| | - Yi Cao
- Department of Experimental Surgery-Cancer Metastasis, Medical Faculty Mannheim, Ruprecht Karls University, Mannheim, 68167, Germany
| | - Dong Tang
- Department of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Institute of General Surgery-Yangzhou, Yangzhou University, Yangzhou, 225000, PR China
| | - Gang Ye
- Department of General Surgery, Jiangdu People's Hospital of Yangzhou, Yangzhou, 225200, PR China
| | - Hongbo Li
- Department of General Surgery, Jiangdu People's Hospital of Yangzhou, Yangzhou, 225200, PR China.
| | - Daorong Wang
- Department of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Institute of General Surgery-Yangzhou, Yangzhou University, Yangzhou, 225000, PR China
| |
Collapse
|
13
|
Lu Y, Yu Y, Liu F, Han Y, Xue H, Sun X, Jiang Y, Tian Z. LINC00511-dependent inhibition of IL-24 contributes to the oncogenic role of HNF4α in colorectal cancer. Am J Physiol Gastrointest Liver Physiol 2021; 320:G338-G350. [PMID: 33052062 DOI: 10.1152/ajpgi.00243.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocyte nuclear factor 4 α (HNF4α) is an important transcription factor that acts as a pro-proliferative mediator during tumorigenesis, yet its function in colorectal cancer (CRC) remain unclear. Hence, this study aims to explore roles that HNF4α plays in the CRC development. RNA quantification analysis was conducted to characterize the expression pattern of long intergenic noncoding RNA 00511 (LINC00511)/HNF4α/IL-24 in CRC tissues and cell lines. Using gain- and loss-of-function approaches, effects of HNF4α/LINC00511/IL-24 axis on biological processes such as proliferative, migrating, invading, apoptotic, and tumorigenic functions of CRC cells were evaluated. We further identified the interactions among HNF4α/LINC00511/EZH2/IL-24 using RNA binding protein immunoprecipitation, RNA pull-down along with chromatin immunoprecipitation (ChIP). LINC00511 was an upregulated lncRNA in CRC tissues and cells, which played an oncogenic role by strengthening the malignant phenotypes of CRC cells. LINC00511 downregulated IL-24 expression by interacting with EZH2. HNF4α could enhance LINC00511 transcription in an epigenetic manner, which finally accelerated cancer progression and tumorigenesis through LINC00511-mediated inhibition of IL-24. Those data together demonstrated the contribution of HNF4α to the progression of CRC through mediating the LINC00511/EZH2/IL-24 axis. Hence, our study provides a promising therapeutic target for CRC.NEW & NOTEWORTHY Our findings on the roles of hepatocyte nuclear factor 4 α/long intergenic noncoding RNA 00511/IL-24 axis provide new insights into the CRC and offer potential targets for translational applications.
Collapse
Affiliation(s)
- Yanyan Lu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yanan Yu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Fuguo Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yue Han
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Huiguang Xue
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Xueguo Sun
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yueping Jiang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| |
Collapse
|
14
|
Qian B, Yao Z, Yang Y, Li N, Wang Q. Downregulation of SDCBP inhibits cell proliferation and induces apoptosis by regulating PI3K/AKT/mTOR pathway in gastric carcinoma. Biotechnol Appl Biochem 2021; 69:240-247. [PMID: 33432665 DOI: 10.1002/bab.2103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/06/2021] [Indexed: 01/04/2023]
Abstract
Syndecan-binding protein (SDCBP) has been reported to critically process a core role in tumorigenesis. This study was conducted to characterize a novel regulatory network of SDCBP in gastric carcinoma (GC) cells. Our findings indicated that overexpression of SDCBP promoted the proliferation of GC cell and increased proliferating cell nuclear antigen (PCNA) expression. Moreover, the overexpression of SDCBP suppressed the apoptosis of GC cell along with a decrease of Bax/Bcl-2 ratio and induction of PI3K/AKT/mTOR activation. However, knockdown of SDCBP exhibited opposed effects on GC cells. Furthermore, silencing SDCBP significantly inhibited GC cell viability and PCNA expression accompanied with the upregulated cell apoptosis and Bax/Bcl-2 ratio, which was regulated by PI3K/AKT/mTOR signaling pathway. And it was further determined that PI3K inhibitor LY294002, AKT inhibitor Torin1, and mTOR inhibitor MK-2206 suppressed the apoptosis. In conclusion, SDCBP promotes the growth ability of GC by inducing the PCNA expression and inhibiting GC cell apoptosis via inactivation of the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Bo Qian
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Zhiheng Yao
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Yang Yang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Na Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Qiao Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
15
|
Cardano M, Tribioli C, Prosperi E. Targeting Proliferating Cell Nuclear Antigen (PCNA) as an Effective Strategy to Inhibit Tumor Cell Proliferation. Curr Cancer Drug Targets 2020; 20:240-252. [PMID: 31951183 DOI: 10.2174/1568009620666200115162814] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 12/20/2022]
Abstract
Targeting highly proliferating cells is an important issue for many types of aggressive tumors. Proliferating Cell Nuclear Antigen (PCNA) is an essential protein that participates in a variety of processes of DNA metabolism, including DNA replication and repair, chromatin organization and transcription and sister chromatid cohesion. In addition, PCNA is involved in cell survival, and possibly in pathways of energy metabolism, such as glycolysis. Thus, the possibility of targeting this protein for chemotherapy against highly proliferating malignancies is under active investigation. Currently, approaches to treat cells with agents targeting PCNA rely on the use of small molecules or on peptides that either bind to PCNA, or act as a competitor of interacting partners. Here, we describe the status of the art in the development of agents targeting PCNA and discuss their application in different types of tumor cell lines and in animal model systems.
Collapse
Affiliation(s)
- Miriana Cardano
- Istituto di Genetica Molecolare del C.N.R. "Luca Cavalli-Sforza", Pavia- 27100, Italy
| | - Carla Tribioli
- Istituto di Genetica Molecolare del C.N.R. "Luca Cavalli-Sforza", Pavia- 27100, Italy
| | - Ennio Prosperi
- Istituto di Genetica Molecolare del C.N.R. "Luca Cavalli-Sforza", Pavia- 27100, Italy
| |
Collapse
|
16
|
Ben-Shmuel A, Biber G, Barda-Saad M. Unleashing Natural Killer Cells in the Tumor Microenvironment-The Next Generation of Immunotherapy? Front Immunol 2020; 11:275. [PMID: 32153582 PMCID: PMC7046808 DOI: 10.3389/fimmu.2020.00275] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence of immunotherapy for cancer treatment bears considerable clinical promise. Nevertheless, many patients remain unresponsive, acquire resistance, or suffer dose-limiting toxicities. Immune-editing of tumors assists their escape from the immune system, and the tumor microenvironment (TME) induces immune suppression through multiple mechanisms. Immunotherapy aims to bolster the activity of immune cells against cancer by targeting these suppressive immunomodulatory processes. Natural Killer (NK) cells are a heterogeneous subset of immune cells, which express a diverse array of activating and inhibitory germline-encoded receptors, and are thus capable of directly targeting and killing cancer cells without the need for MHC specificity. Furthermore, they play a critical role in triggering the adaptive immune response. Enhancing the function of NK cells in the context of cancer is therefore a promising avenue for immunotherapy. Different NK-based therapies have been evaluated in clinical trials, and some have demonstrated clinical benefits, especially in the context of hematological malignancies. Solid tumors remain much more difficult to treat, and the time point and means of intervention of current NK-based treatments still require optimization to achieve long term effects. Here, we review recently described mechanisms of cancer evasion from NK cell immune surveillance, and the therapeutic approaches that aim to potentiate NK function. Specific focus is placed on the use of specialized monoclonal antibodies against moieties on the cancer cell, or on both the tumor and the NK cell. In addition, we highlight newly identified mechanisms that inhibit NK cell activity in the TME, and describe how biochemical modifications of the TME can synergize with current treatments and increase susceptibility to NK cell activity.
Collapse
Affiliation(s)
- Aviad Ben-Shmuel
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Guy Biber
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Mira Barda-Saad
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
17
|
Shi J, Guan X, Zhan F, Liu C, Li Z, Yao Y, Wang B, Lou C, Zhang Y. CSN6 expression is associated with pancreatic cancer progression and predicts poor prognosis. Cancer Biol Ther 2019; 20:1290-1299. [PMID: 31311398 DOI: 10.1080/15384047.2019.1632143] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Constitutive photomorphogenesis 9 (COP9) signalosome 6 (CSN6) plays an essential role in tumor development. The present study aims to demonstrate that CSN6 is an important biomarker and has prognostic value for patients with pancreatic ductal adenocarcinoma (PDAC). We analyzed CSN6 expression levels in PDAC and adjacent non-cancerous tissues using immunohistochemistry (IHC) and quantitative real-time PCR (qPCR) analysis. We found that CSN6 was highly expressed in PDAC tissues, contrasting to adjacent non-cancerous tissues. Interestingly, CSN6 expression was positively associated with proliferating cell nuclear antigen (PCNA) expression. Further investigation indicated that CSN6 knockdown significantly suppressed the proliferation of PDAC cells and decreased the expression levels of PCNA, while CSN6 overexpression increased the proliferation, as well as the expression levels of PCNA in PDAC cells. Furthermore, a χ2 test indicated that the expression of CSN6 in PDAC tissues was markedly associated with tumor infiltration and serum carbohydrate antigen 19-9 levels. In addition, univariate and multivariate analyses showed that CSN6 levels were significantly correlated with poor clinical outcomes of patients with PDAC. Kaplan-Meier analysis showed that patients with high expression of CSN6 had shorter overall survival. These results suggest that the expression of CSN6 correlates with the progression of PDAC, resulting in poor prognosis. Thus, CSN6 may play a significant role in the development of PDAC and is a potential target to prevent and treat PDAC.
Collapse
Affiliation(s)
- Jiaqi Shi
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital , Harbin , Heilongjiang Province , P. R. China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences , Harbin , P. R. China
| | - Xin Guan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital , Harbin , Heilongjiang Province , P. R. China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences , Harbin , P. R. China
| | - Fei Zhan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital , Harbin , Heilongjiang Province , P. R. China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences , Harbin , P. R. China
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital , Harbin , Heilongjiang Province , P. R. China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences , Harbin , P. R. China
| | - Zhiwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital , Harbin , Heilongjiang Province , P. R. China
| | - Yuanfei Yao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital , Harbin , Heilongjiang Province , P. R. China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences , Harbin , P. R. China
| | - Bojun Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital , Harbin , Heilongjiang Province , P. R. China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences , Harbin , P. R. China
| | - Changjie Lou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital , Harbin , Heilongjiang Province , P. R. China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital , Harbin , Heilongjiang Province , P. R. China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences , Harbin , P. R. China
| |
Collapse
|
18
|
Kundu K, Ghosh S, Sarkar R, Edri A, Brusilovsky M, Gershoni-Yahalom O, Yossef R, Shemesh A, Soria JC, Lazar V, Joshua BZ, Campbell KS, Elkabets M, Porgador A. Inhibition of the NKp44-PCNA Immune Checkpoint Using a mAb to PCNA. Cancer Immunol Res 2019; 7:1120-1134. [PMID: 31164357 DOI: 10.1158/2326-6066.cir-19-0023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/09/2019] [Accepted: 05/30/2019] [Indexed: 02/05/2023]
Abstract
mAb-based blocking of the immune checkpoints involving the CTLA4-B7 and PD1-PDL1 inhibitory axes enhance T-cell-based adaptive immune responses in patients with cancer. We show here that antitumor responses by natural killer (NK) cells can be enhanced by a checkpoint-blocking mAb, 14-25-9, which we developed against proliferating cell nuclear antigen (PCNA). PCNA is expressed on the surface of cancer cells and acts as an inhibitory ligand for the NK-cell receptor, NKp44-isoform1. We tested for cytoplasmic- and membrane-associated PCNA by FACS- and ImageStream-based staining of cell lines and IHC of human cancer formalin fixed, paraffin embedded tissues. The mAb, 14-25-9, inhibited binding of chimeric NKp44 receptor to PCNA and mostly stained the cytoplasm and membrane of tumor cells, whereas commercial antibody (clone PC10) stained nuclear PCNA. NK functions were measured using ELISA-based IFNγ secretion assays and FACS-based killing assays. The NK92-NKp44-1 cell line and primary human NK cells showed increased IFNγ release upon coincubation with mAb 14-25-9 and various solid tumor cell lines and leukemias. Treatment with 14-25-9 also increased NK cytotoxic activity. In vivo efficacy was evaluated on patient-derived xenografts (PDX)-bearing NSG mice. In PDX-bearing mice, intravenous administration of mAb 14-25-9 increased degranulation (CD107a expression) of intratumorally injected patient autologous or allogeneic NK cells, as well as inhibited tumor growth when treated long term. Our study describes a mAb against the NKp44-PCNA innate immune checkpoint that can enhance NK-cell antitumor activity both in vitro and in vivo.
Collapse
Affiliation(s)
- Kiran Kundu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Susmita Ghosh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Rhitajit Sarkar
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Avishay Edri
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Michael Brusilovsky
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Orly Gershoni-Yahalom
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Rami Yossef
- Surgery Branch, NCI, NIH, Bethesda, Maryland
| | - Avishai Shemesh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | | | - Vladimir Lazar
- Worldwide Innovative Network (WIN) Association - WIN Consortium, Villejuif, France
| | - Ben-Zion Joshua
- Department of Otolaryngology-Head and Neck Surgery, Soroka Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kerry S Campbell
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel. .,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
19
|
Girard P, Charles J, Cluzel C, Degeorges E, Manches O, Plumas J, De Fraipont F, Leccia MT, Mouret S, Chaperot L, Aspord C. The features of circulating and tumor-infiltrating γδ T cells in melanoma patients display critical perturbations with prognostic impact on clinical outcome. Oncoimmunology 2019; 8:1601483. [PMID: 31413911 DOI: 10.1080/2162402x.2019.1601483] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 02/08/2023] Open
Abstract
γδT cells hold a pivotal role in tumor immunosurveillance through their prompt activation and cytokine secretion, their ability to kill tumor cells in an Human Leukocyte Antigen (HLA)-unrestricted manner, and their combination of features of both innate and adaptive immunity. These unique properties and functional plasticity render them very attractive both as targets and vectors for cancer immunotherapy. Yet, these potent and fascinating antitumor effectors have not been extensively explored in melanoma. We provided here a detailed investigation of the phenotypic and functional properties of circulating and tumor-infiltrating γδT cells in melanoma patients, and their impact on clinical evolution. High proportions of circulating- and tumor-infiltrating γδT and δ2+ subset were associated with better clinical outcome. We reported however that circulating and tumor-infiltrating γδT cells from melanoma patients displayed an altered expression of NCR, KIR, and immune checkpoints, and identified NKp44, PD1, 41BB/41BBL, TIM3, and LAG3 as crucial checkpoints allowing immune escape and tumor progression. Notably, melanoma drastically impaired the ability of γδT cells to exhibit activation molecules, secrete cytokines, and display cytotoxicity toward melanoma in response to stimulation with phosphoantigens. It drove them toward regulatory and Th17 profiles associated with poor clinical outcomes. Our study highlights that melanoma hijacked γδT cells to escape from immune control, and revealed that circulating and tumor-infiltrating γδT cell features are promising potential biomarkers of clinical evolution. Such understanding of the physiopathology of γδT cells may help designing new therapeutic approaches exploiting the antitumor potential of γδT cells while counteracting their skewing by tumors to improve patient outcomes.
Collapse
Affiliation(s)
- Pauline Girard
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Julie Charles
- University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,Dermatology clinic, Grenoble University Hospital, Grenoble, France
| | - Camille Cluzel
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Emmanuelle Degeorges
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Olivier Manches
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Joel Plumas
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,pDCline Pharma, Grenoble, France
| | - Florence De Fraipont
- University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,Department of Biochemistry of Cancers and Biotherapies, Grenoble University Hospital, Grenoble, France
| | - Marie-Therese Leccia
- University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,Dermatology clinic, Grenoble University Hospital, Grenoble, France
| | - Stephane Mouret
- University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,Dermatology clinic, Grenoble University Hospital, Grenoble, France
| | - Laurence Chaperot
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Caroline Aspord
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| |
Collapse
|
20
|
Parodi M, Favoreel H, Candiano G, Gaggero S, Sivori S, Mingari MC, Moretta L, Vitale M, Cantoni C. NKp44-NKp44 Ligand Interactions in the Regulation of Natural Killer Cells and Other Innate Lymphoid Cells in Humans. Front Immunol 2019; 10:719. [PMID: 31024551 PMCID: PMC6465645 DOI: 10.3389/fimmu.2019.00719] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
Natural Killer (NK) cells are potent cytotoxic cells belonging to the family of Innate Lymphoid Cells (ILCs). Their most characterized effector functions are directed to the control of aberrant cells in the body, including both transformed and virus-infected cells. NK cell-mediated recognition of abnormal cells primarily occurs through receptor-ligand interactions, involving an array of inhibitory and activating NK receptors and different types of ligands expressed on target cells. While most of the receptors have become known over many years, their respective ligands were only defined later and their impressive complexity has only recently become evident. NKp44, a member of Natural Cytotoxicity Receptors (NCRs), is an activating receptor playing a crucial role in most functions exerted by activated NK cells and also by other NKp44+ immune cells. The large and heterogeneous panel of NKp44 ligands (NKp44L) now includes surface expressed glycoproteins and proteoglycans, nuclear proteins that can be exposed outside the cell, and molecules that can be either released in the extracellular space or carried in extracellular vesicles. Recent findings have extended our knowledge on the nature of NKp44L to soluble plasma glycoproteins, such as secreted growth factors or extracellular matrix (ECM)-derived glycoproteins. NKp44L are induced upon tumor transformation or viral infection but may also be expressed in normal cells and tissues. In addition, NKp44-NKp44L interactions are involved in the crosstalk between NK cells and different innate and adaptive immune cell types. NKp44 expression in different ILCs located in tissues further extends the potential role of NKp44-NKp44L interactions.
Collapse
Affiliation(s)
- Monica Parodi
- Immunology Operative Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Herman Favoreel
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Silvia Gaggero
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Simona Sivori
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Maria Cristina Mingari
- Immunology Operative Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Massimo Vitale
- Immunology Operative Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Claudia Cantoni
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.,Laboratory of Clinical and Experimental Immunology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
21
|
Pelizaro BI, Braga FC, Crispim BDA, de Barros LGML, Pessatto LR, Oliveira EJT, Vani JM, de Souza AP, Grisolia AB, Antoniolli-Silva ACMB, de Lima DP, Dos Santos Jaques JA, Beatriz A, Oliveira RJ. Assessment of acute toxicity and cytotoxicity of fluorescent markers produced by cardanol and glycerol, which are industrial waste, to different biological models. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:9193-9202. [PMID: 30719661 DOI: 10.1007/s11356-019-04376-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/24/2019] [Indexed: 06/09/2023]
Abstract
The amphyphylic triazoanilines recently synthesized 1-(4-(3-aminophenyl)-1H-1,2,3- triazole-1-yl)-3-(3-pentadecylphenoxy)propan-2-ol (1) and 1-(4-(4-aminophenyl)-1H- 1,2,3-triazole-1-yl)-3-(3-pentadecylphenoxy)propan-2-ol (2), synthesized from cardanol and glycerol, have photophysical properties which allow their use in the development of fluorescent biomarkers with applicability in the biodiesel quality control. Based on this, the present research evaluated the toxic effects of both compounds in different biological models through the investigation of survival and mortality percentages as a measure of acute toxicity on Daphnia similis and Oreochromis niloticus, larvicidal assay against Aedes aegypti, and cytotoxic activity on mammary cells. Results demonstrate that these triazoanilines 1 and 2 have shown low acute toxicity to the biological models investigated in this study up to the following concentrations: 4.0 mg L-1 (D. similis), 4.0 mg L-1 (A. aegypti larvae), 1.0 mg L-1 (O. niloticus), and 1.0 mg mL-1 (mammary cells). This fact suggests the potential for safe use of compounds 1 and 2 as fluorescent markers for the monitoring of biodiesel quality, even in the case of environmental exposure. Besides all of that, the reuse of cardanol and glycerol, both industrial wastes, favors the maintenance of environmental health and is in agreement with the assumptions of green chemistry. Graphical abstract ᅟ.
Collapse
Affiliation(s)
- Bruno Ivo Pelizaro
- Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Brazilian Hospital Services Company (EBSERH), Campo Grande, Mato Grosso do Sul, Brazil
- Master's Program in Pharmacy, Faculty of Pharmaceutical Sciences, Food and Nutrition - FACFAN, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Felipe Camargo Braga
- Chemistry Institute - INQUI, SINTMOL Laboratory, Federal University of Mato Grosso do Sul, Avenue Senador Filinto Müller, 1555 - Universitário, Campo Grande, Mato Grosso do Sul, 79074-460, Brazil
| | - Bruno do Amaral Crispim
- Laboratory of Ecotoxicology and Genotoxicity (LECOGEN), Faculty of Biological and Environmental Sciences - FCBA, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil
| | - Luiz Guilherme Maiolino Lacerda de Barros
- Laboratory of Environmental Quality, Faculty of Engineering Architecture and Urbanism and Geography - FAENG, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Lucas Roberto Pessatto
- Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Brazilian Hospital Services Company (EBSERH), Campo Grande, Mato Grosso do Sul, Brazil
- Graduate Programme in Genetics and Molecular Biology, Department of General Biology, State University of Londrina (UEL), Londrina, Paraná, Brazil
| | - Edwin José Torres Oliveira
- Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Brazilian Hospital Services Company (EBSERH), Campo Grande, Mato Grosso do Sul, Brazil
- Graduate Programme in Genetics and Molecular Biology, Department of General Biology, State University of Londrina (UEL), Londrina, Paraná, Brazil
| | - Juliana Miron Vani
- Graduate Programme in Health and Development in the Central-West Region, School of Medicine (FAMED) "Dr. Hélio Mandetta", Federal University of Mato Grosso do Sul (UFMS), Cidade Universitária, S/N., Campo Grande, MS, 79070-900, Brazil
| | - Antonio Pancrácio de Souza
- Biosciences Institute- INBIO, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Alexeia Barufatti Grisolia
- Laboratory of Ecotoxicology and Genotoxicity (LECOGEN), Faculty of Biological and Environmental Sciences - FCBA, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil
| | - Andréia Conceição Milan Brochado Antoniolli-Silva
- Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Brazilian Hospital Services Company (EBSERH), Campo Grande, Mato Grosso do Sul, Brazil
- Graduate Programme in Health and Development in the Central-West Region, School of Medicine (FAMED) "Dr. Hélio Mandetta", Federal University of Mato Grosso do Sul (UFMS), Cidade Universitária, S/N., Campo Grande, MS, 79070-900, Brazil
| | - Dênis Pires de Lima
- Chemistry Institute - INQUI, SINTMOL Laboratory, Federal University of Mato Grosso do Sul, Avenue Senador Filinto Müller, 1555 - Universitário, Campo Grande, Mato Grosso do Sul, 79074-460, Brazil
| | - Jeandre Augusto Dos Santos Jaques
- Master's Program in Pharmacy, Faculty of Pharmaceutical Sciences, Food and Nutrition - FACFAN, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Adilson Beatriz
- Chemistry Institute - INQUI, SINTMOL Laboratory, Federal University of Mato Grosso do Sul, Avenue Senador Filinto Müller, 1555 - Universitário, Campo Grande, Mato Grosso do Sul, 79074-460, Brazil.
| | - Rodrigo Juliano Oliveira
- Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Brazilian Hospital Services Company (EBSERH), Campo Grande, Mato Grosso do Sul, Brazil.
- Graduate Programme in Genetics and Molecular Biology, Department of General Biology, State University of Londrina (UEL), Londrina, Paraná, Brazil.
- Graduate Programme in Health and Development in the Central-West Region, School of Medicine (FAMED) "Dr. Hélio Mandetta", Federal University of Mato Grosso do Sul (UFMS), Cidade Universitária, S/N., Campo Grande, MS, 79070-900, Brazil.
| |
Collapse
|