1
|
Hua Y, Jiang P, Dai C, Li M. Extracellular vesicle autoantibodies. J Autoimmun 2024; 149:103322. [PMID: 39341173 DOI: 10.1016/j.jaut.2024.103322] [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: 07/16/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024]
Abstract
Autoantibodies are immunoglobulin proteins produced by autoreactive B cells responding to self-antigens. Extracellular vesicles (EVs) are membranous structures released by almost all types of cells and extensively distributed in various biological fluids. Studies have indicated that EVs loaded with self-antigens not only play important roles in antigen presentation and autoantibody production but can also form functional immune complexes with autoantibodies (termed EV autoantibodies). While numerous papers have summarized the production and function of pathogenic autoantibodies in diseases, especially autoimmune diseases, reviews on EV autoantibodies are rare. In this review, we outline the existing knowledge about EVs, autoantibodies, and EV antigens, highlighting the formation of EV autoantibodies and their functions in autoimmune diseases and cancers. In conclusion, EV autoantibodies may be involved in the occurrence of disease(s) and also serve as potential non-invasive markers that could help in the diagnosis and/or prognosis of disease. Additional studies designed to define in more detail the molecular characteristics of EV autoantibodies and their contribution to disease are recommended.
Collapse
Affiliation(s)
- Yan Hua
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China; Core Unit of National Clinical Research Center for Laboratory Medicine of China, Hefei, Anhui, 230001, China
| | - Panpan Jiang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Chunyang Dai
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China; Core Unit of National Clinical Research Center for Laboratory Medicine of China, Hefei, Anhui, 230001, China
| | - Ming Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China; Core Unit of National Clinical Research Center for Laboratory Medicine of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
2
|
Sanadgol N, Amini J, Khalseh R, Bakhshi M, Nikbin A, Beyer C, Zendehdel A. Mitochondrial genome-derived circRNAs: Orphan epigenetic regulators in molecular biology. Mitochondrion 2024; 79:101968. [PMID: 39321951 DOI: 10.1016/j.mito.2024.101968] [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: 05/17/2024] [Revised: 09/02/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Mitochondria are vital for cellular activities, influencing ATP production, Ca2+ signaling, and reactive oxygen species generation. It has been proposed that nuclear genome-derived circular RNAs (circRNAs) play a role in biological processes. For the first time, this study aims to comprehensively explore experimentally confirmed human mitochondrial genome-derived circRNAs (mt-circRNAs) via in-silico analysis. We utilized wide-ranging bioinformatics tools to anticipate their roles in molecular biology, involving miRNA sponging, protein antagonism, and peptide translation. Among five well-characterized mt-circRNAs, SCAR/mc-COX2 stands out as particularly significant with the potential to sponge around 41 different miRNAs, which target several genes mostly involved in endocytosis, MAP kinase, and PI3K-Akt pathways. Interestingly, circMNTND5 and mecciND1 specifically interact with miRNAs through their unique back-splice junction sequence. These exclusively targeted miRNAs (has-miR-5186, 6888-5p, 8081, 924, 672-5p) are predominantly associated with insulin secretion, proteoglycans in cancer, and MAPK signaling pathways. Moreover, all mt-circRNAs intricately affect the P53 pathway through miRNA sequestration. Remarkably, mc-COX2 and circMNTND5 appear to be involved in the RNA's biogenesis by antagonizing AGO1/2, EIF4A3, and DGCR8. All mt-circRNAs engaged with IGF2BP proteins crucial in redox signaling, and except mecciND1, they all potentially generate at least one protein resembling the immunoglobulin heavy chain protein. Given P53's function as a redox-sensitive transcription factor, and insulin's role as a crucial regulator of energy metabolism, their indirect interplay with mt-circRNAs could influence cellular outcomes. However, due to limited attention and infrequent data availability, it is advisable to conduct more thorough investigations to gain a deeper understanding of the functions of mt-circRNA.
Collapse
Affiliation(s)
- Nima Sanadgol
- Institute of Neuroanatomy, RWTH University Hospital Aachen, 52074 Aachen, Germany.
| | - Javad Amini
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, 94149-75516 Bojnurd, Iran
| | - Roghayeh Khalseh
- Institute of Neuroanatomy, RWTH University Hospital Aachen, 52074 Aachen, Germany
| | - Mostafa Bakhshi
- Department of Electrical and Computer Engineering, Kharazmi University, 15719-14911 Tehran, Iran
| | - Arezoo Nikbin
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Golestan University of Medical Sciences, Gorgan, Iran
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH University Hospital Aachen, 52074 Aachen, Germany
| | - Adib Zendehdel
- Institut of Anatomy, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
3
|
Zhang W, Chen X, Chen X, Li J, Wang H, Yan X, Zha H, Ma X, Zhao C, Su M, Hong L, Li P, Ling Y, Zhao W, Xia Y, Li B, Zheng T, Gu J. Fc-Fc interactions and immune inhibitory effects of IgG4: implications for anti-PD-1 immunotherapies. J Immunother Cancer 2024; 12:e009034. [PMID: 38925680 PMCID: PMC11203076 DOI: 10.1136/jitc-2024-009034] [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] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The majority of anti-programmed cell-death 1 (PD-1) monoclonal antibodies (mAbs) use S228P mutation IgG4 as the structural basis to avoid the activation of immune cells or complement. However, little attention has been paid to the Fc-Fc interactions between IgG4 and other IgG Fc fragments that could result in adverse effects. Fc-null IgG1 framework is a potential safer alternative to avoid the undesirable Fc-Fc interactions and Fc receptor binding derived effects observed with IgG4. This study provides a comprehensive evaluation of anti-PD-1 mAbs of these two frameworks. METHODS Trastuzumab and rituximab (both IgG1), wildtype IgG1 and IgG4 were immobilized on nitrocellulose membranes, coated to microplates and biosensor chips, and bound to tumor cells as targets for Fc-Fc interactions. Wildtype IgG1 and IgG4, anti-PD-1 mAb nivolumab (IgG4 S228P), penpulimab (Fc-null IgG1), and tislelizumab (Fc-null IgG4 S228P-R409K) were assessed for their binding reactions to the immobilized IgG proteins and quantitative kinetic data were obtained. To evaluate the effects of the two anti-PD-1 mAbs on immune responses mediated by trastuzumab and rituximab in the context of combination therapy, we employed classic immune models for antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and complement dependent cytotoxicity. Tumor-bearing mouse models, both wildtype and humanized, were used for in vivo investigation. Furthermore, we also examined the effects of IgG1 and IgG4 on diverse immune cell populations RESULTS: Experiments demonstrated that wildtype IgG4 and nivolumab bound to immobilized IgG through Fc-Fc interactions, diminishing antibody-dependent cell-mediated cytotoxicity and phagocytosis reactions. Quantitative analysis of kinetic parameters suggests that nivolumab and wildtype IgG4 exhibit comparable binding affinities to immobilized IgG1 in both non-denatured and denatured states. IgG4 exerted inhibitory effects on various immune cell types. Wildtype IgG4 and nivolumab both promoted tumor growth in wildtype mouse models. Conversely, wildtype IgG1, penpulimab, and tislelizumab did not show similar adverse effects. CONCLUSIONS Fc-null IgG1 represents a safer choice for anti-PD-1 immunotherapies by avoiding both the adverse Fc-Fc interactions and Fc-related immune inhibitory effects of IgG4. Fc-null IgG4 S228P-R409K and Fc-null IgG1 displayed similar structural properties and benefits. This study contributes to the understanding of immunotherapy resistance and the advancement of safer immune therapies for cancer.
Collapse
Affiliation(s)
- Weifeng Zhang
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Xueling Chen
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Xingxing Chen
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Jirui Li
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Hui Wang
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Xiaomiao Yan
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
- Jinxin Research Institute for Reproductive Medicine and Genetics, Xinan Hospital for Maternal and Child Health Care, Chengdu, China
| | - Han Zha
- The People's Hospital of Qijiang District Chongqing, Chongqing, China
| | - Xiaonan Ma
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Chanyuan Zhao
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Meng Su
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Liangli Hong
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Penghao Li
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
- Jinxin Research Institute for Reproductive Medicine and Genetics, Xinan Hospital for Maternal and Child Health Care, Chengdu, China
| | - Yanyu Ling
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Wenhui Zhao
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Yu Xia
- Akeso Biopharma Inc, Zhongshan, China
| | | | - Tianjing Zheng
- Chia Tai Tianqing Pharmaceutical Group Co., LTD, Nanjing, China
| | - Jiang Gu
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
- Jinxin Research Institute for Reproductive Medicine and Genetics, Xinan Hospital for Maternal and Child Health Care, Chengdu, China
| |
Collapse
|
4
|
Chen X, Ke H, Li W, Yin L, Chen W, Chen T, Wu Y, Qiu J, Feng W. Structural basis for the recognition of IFNAR1 by the humanized therapeutic monoclonal antibody QX006N for the treatment of systemic lupus erythematosus. Int J Biol Macromol 2024; 268:131721. [PMID: 38649079 DOI: 10.1016/j.ijbiomac.2024.131721] [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: 02/02/2024] [Revised: 04/10/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Interferon (IFN) alpha/beta receptor 1 (IFNAR1) is indispensable for antiviral responses and the immune regulation. Dysregulation of the IFNAR1-mediaetd signaling pathways leads to deleterious autoimmune diseases such as systemic lupus erythematosus (SLE). QX006N, a humanized therapeutic monoclonal antibody, specifically targets human IFNAR1 and is in the clinical trial phase for treating SLE, but the molecular mechanism underlying the QX006N-mediated recognition of IFNAR1 remains unclear. Here, we report the high neutralization activities of QX006N against IFNAR1-mediated signal transduction. Meanwhile, we determine the structures of the fragment antigen-binding domain (Fab) of QX006N (QX006N-Fab) and QX006N-Fab in complex with the subdomains 1-3 of IFNAR1 (IFNAR1-SD123) at 2.87 Å and 2.68 Å resolutions, respectively. In the structure of the QX006N-Fab/IFNAR1-SD123 complex, QX006N-Fab only recognizes the SD3 subdomain of IFNAR1 by the hydrophobic, hydrogen-bonding and electrostatic interactions. Compared with the structure of the IFN/IFNAR1/IFNAR2 complex, the binding of QX006N-Fab to IFNAR1-SD3 blocks its association with IFN due to steric hindrance, which inhibits the IFN/IFNAR1/IFNAR2 complex formation for signal transduction. The results of this study provide the structural evidence for the specific targeting of IFNAR1 by the therapeutic antibody QX006N and pave the way for the rational design of antibody drugs to combat IFNAR1-related autoimmune diseases.
Collapse
MESH Headings
- Receptor, Interferon alpha-beta/metabolism
- Receptor, Interferon alpha-beta/chemistry
- Lupus Erythematosus, Systemic/drug therapy
- Lupus Erythematosus, Systemic/immunology
- Humans
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Protein Binding
- Models, Molecular
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/immunology
- Signal Transduction/drug effects
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Xiaorong Chen
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Huimin Ke
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China.
| | - Wei Li
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Lu Yin
- Qyuns Therapeutics Co., Ltd., Taizhou 225300, China
| | - Wei Chen
- Qyuns Therapeutics Co., Ltd., Taizhou 225300, China
| | - Tao Chen
- Qyuns Therapeutics Co., Ltd., Taizhou 225300, China
| | - Yiliang Wu
- Qyuns Therapeutics Co., Ltd., Taizhou 225300, China
| | - Jiwan Qiu
- Qyuns Therapeutics Co., Ltd., Taizhou 225300, China.
| | - Wei Feng
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
5
|
Szűcs D, Monostori T, Miklós V, Páhi ZG, Póliska S, Kemény L, Veréb Z. Licensing effects of inflammatory factors and TLR ligands on the regenerative capacity of adipose-derived mesenchymal stem cells. Front Cell Dev Biol 2024; 12:1367242. [PMID: 38606318 PMCID: PMC11007080 DOI: 10.3389/fcell.2024.1367242] [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: 01/08/2024] [Accepted: 03/15/2024] [Indexed: 04/13/2024] Open
Abstract
Introduction: Adipose tissue-derived mesenchymal stem cells are promising contributors to regenerative medicine, exhibiting the ability to regenerate tissues and modulate the immune system, which is particularly beneficial for addressing chronic inflammatory ulcers and wounds. Despite their inherent capabilities, research suggests that pretreatment amplifies therapeutic effectiveness. Methods: Our experimental design exposed adipose-derived mesenchymal stem cells to six inflammatory factors for 24 h. We subsequently evaluated gene expression and proteome profile alterations and observed the wound closure rate post-treatment. Results: Specific pretreatments, such as IL-1β, notably demonstrated an accelerated wound-healing process. Analysis of gene and protein expression profiles revealed alterations in pathways associated with tissue regeneration. Discussion: This suggests that licensed cells exhibit potentially higher therapeutic efficiency than untreated cells, shedding light on optimizing regenerative strategies using adipose tissue-derived stem cells.
Collapse
Affiliation(s)
- Diána Szűcs
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
- Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Szeged, Hungary
| | - Tamás Monostori
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
- Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Szeged, Hungary
| | | | - Zoltán G. Páhi
- Genome Integrity and DNA Repair Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), University of Szeged, Szeged, Hungary
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Szilárd Póliska
- Genomic Medicine and Bioinformatics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Lajos Kemény
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
- Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine-USz Skin Research Group, University of Szeged, Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
- Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Szeged, Hungary
- Biobank, University of Szeged, Szeged, Hungary
| |
Collapse
|
6
|
Yan K, Liu Q, Huang R, Jiang Y, Bian Z, Li S, Li L, Shen F, Tsuneyama K, Zhang Q, Lian Z, Guan H, Xu B. Spatial transcriptomics reveals prognosis-associated cellular heterogeneity in the papillary thyroid carcinoma microenvironment. Clin Transl Med 2024; 14:e1594. [PMID: 38426403 PMCID: PMC10905537 DOI: 10.1002/ctm2.1594] [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: 08/02/2023] [Revised: 01/28/2024] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is the most common malignant endocrine tumour, and its incidence and prevalence are increasing considerably. Cellular heterogeneity in the tumour microenvironment is important for PTC prognosis. Spatial transcriptomics is a powerful technique for cellular heterogeneity study. METHODS In conjunction with a clinical pathologist identification method, spatial transcriptomics was employed to characterise the spatial location and RNA profiles of PTC-associated cells within the tissue sections. The spatial RNA-clinical signature genes for each cell type were extracted and applied to outlining the distribution regions of specific cells on the entire section. The cellular heterogeneity of each cell type was further revealed by ContourPlot analysis, monocle analysis, trajectory analysis, ligand-receptor analysis and Gene Ontology enrichment analysis. RESULTS The spatial distribution region of tumour cells, typical and atypical follicular cells (FCs and AFCs) and immune cells were accurately and comprehensively identified in all five PTC tissue sections. AFCs were identified as a transitional state between FCs and tumour cells, exhibiting a higher resemblance to the latter. Three tumour foci were shared among all patients out of the 13 observed. Notably, tumour foci No. 2 displayed elevated expression levels of genes associated with lower relapse-free survival in PTC patients. We discovered key ligand-receptor interactions, including LAMB3-ITGA2, FN1-ITGA3 and FN1-SDC4, involved in the transition of PTC cells from FCs to AFCs and eventually to tumour cells. High expression of these patterns correlated with reduced relapse-free survival. In the tumour immune microenvironment, reduced interaction between myeloid-derived TGFB1 and TGFBR1 in tumour focus No. 2 contributed to tumourigenesis and increased heterogeneity. The spatial RNA-clinical analysis method developed here revealed prognosis-associated cellular heterogeneity in the PTC microenvironment. CONCLUSIONS The occurrence of tumour foci No. 2 and three enhanced ligand-receptor interactions in the AFC area/tumour foci reduced the relapse-free survival of PTC patients, potentially leading to improved prognostic strategies and targeted therapies for PTC patients.
Collapse
Affiliation(s)
- Kai Yan
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Qing‐Zhi Liu
- Chronic Disease LaboratoryInstitutes for Life SciencesSouth China University of TechnologyGuangzhouChina
| | - Rong‐Rong Huang
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Yi‐Hua Jiang
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and ApplicationGuangzhouChina
| | - Zhen‐Hua Bian
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouChina
| | - Si‐Jin Li
- Department of Thyroid SurgeryGuangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Liang Li
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Fei Shen
- Department of Thyroid SurgeryGuangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory MedicineInstitute of Biomedical SciencesTokushima University Graduate SchoolTokushimaJapan
| | - Qing‐Ling Zhang
- Department of PathologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Zhe‐Xiong Lian
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Haixia Guan
- Department of EndocrinologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Bo Xu
- Department of Thyroid SurgeryGuangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| |
Collapse
|
7
|
Augustine TN, Buthelezi S, Pather K, Xulu KR, Stoychev S. Secretomics reveals hormone-therapy of breast cancer may induce survival by facilitating hypercoagulation and immunomodulation in vitro. Sci Rep 2024; 14:1486. [PMID: 38233507 PMCID: PMC10794708 DOI: 10.1038/s41598-023-49755-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024] Open
Abstract
Tumour cell haematogenous dissemination is predicated on molecular changes that enhance their capacity for invasion and preparation of the pre-metastatic niche. It is increasingly evident that platelets play an essential role in this transformation. The systemic nature of signalling molecules and extravascular factors that participate in mediating platelet-tumour cell interactions led to the development of an in vitro co-culture using whole blood and breast tumour cells, allowing us to decipher the impact of hormone-therapy on tumour cells and associated changes in the plasma proteome. Using mass spectrometry, we determined dysregulation of proteins associated with maintaining an invasive tumour phenotype. Tumour changes in genes associated with EMT and survival were documented. This is postulated to be induced via tumour cell interactions with the coagulatory and immune systems. Results highlight tumour cell adaptability to both treatment and blood resulting in a pro-tumorigenic response and a hypercoagulatory state. We illustrate that the breast cancer cell secretome can be altered by hormone-therapy, subject to the tumour subphenotype and linked to platelet activation. More sophisticated co-culture systems are required to recapitulate these interactions to better understand tumorigenesis. Moreover, deeper plasma profiling, using abundant protein depleted and/or vesicle enriched strategies, will likely reveal additional secretory proteins related to tumour cell-platelet interactions.
Collapse
Affiliation(s)
- Tanya N Augustine
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Sindisiwe Buthelezi
- Department of Biosciences, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Kyrtania Pather
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kutlwano R Xulu
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stoyan Stoychev
- Department of Biosciences, Council for Scientific and Industrial Research, Pretoria, South Africa.
| |
Collapse
|
8
|
Thapa R, Gupta G, Bhat AA, Almalki WH, Alzarea SI, Kazmi I, Saleem S, Khan R, Altwaijry N, Dureja H, Singh SK, Dua K. A review of Glycogen Synthase Kinase-3 (GSK3) inhibitors for cancers therapies. Int J Biol Macromol 2023; 253:127375. [PMID: 37839597 DOI: 10.1016/j.ijbiomac.2023.127375] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
The intricate molecular pathways governing cancer development and progression have spurred intensive investigations into novel therapeutic targets. Glycogen Synthase Kinase-3 (GSK3), a complex serine/threonine kinase, has emerged as a key player with intricate roles in various cellular processes, including cell proliferation, differentiation, apoptosis, and metabolism. Harnessing GSK3 inhibitors as potential candidates for cancer therapy has garnered significant interest due to their ability to modulate key signalling pathways that drive oncogenesis. The review encompasses a thorough examination of the molecular mechanisms underlying GSK3's involvement in cancer progression, shedding light on its interaction with critical pathways such as Wnt/β-catenin, PI3K/AKT, and NF-κB. Through these interactions, GSK3 exerts influence over tumour growth, invasion, angiogenesis, and metastasis, rendering it an attractive target for therapeutic intervention. The discussion includes preclinical and clinical studies, showcasing the inhibitors efficacy across a spectrum of cancer types, including pancreatic, ovarian, lung, and other malignancies. Insights from recent studies highlight the potential synergistic effects of combining GSK3 inhibitors with conventional chemotherapeutic agents or targeted therapies, opening avenues for innovative combinatorial approaches. This review provides a comprehensive overview of the current state of research surrounding GSK3 inhibitors as promising agents for cancer treatment.
Collapse
Affiliation(s)
- Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India.
| | - Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shakir Saleem
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Ruqaiyah Khan
- Department of Basic Health Sciences, Deanship of Preparatory Year for the Health Colleges, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Najla Altwaijry
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Harish Dureja
- Faculty of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
9
|
Lefrère H, Moore K, Floris G, Sanders J, Seignette IM, Bismeijer T, Peters D, Broeks A, Hooijberg E, Van Calsteren K, Neven P, Warner E, Peccatori FA, Loibl S, Maggen C, Han SN, Jerzak KJ, Annibali D, Lambrechts D, de Visser KE, Wessels L, Lenaerts L, Amant F. Poor Outcome in Postpartum Breast Cancer Patients Is Associated with Distinct Molecular and Immunologic Features. Clin Cancer Res 2023; 29:3729-3743. [PMID: 37449970 PMCID: PMC10502474 DOI: 10.1158/1078-0432.ccr-22-3645] [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: 11/23/2022] [Revised: 03/23/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE Patients with postpartum breast cancer diagnosed after cessation of breastfeeding (postweaning, PP-BCPW) have a particularly poor prognosis compared with patients diagnosed during lactation (PP-BCDL), or to pregnant (Pr-BC) and nulliparous (NP-BC) patients, regardless of standard prognostic characteristics. Animal studies point to a role of the involution process in stimulation of tumor growth in the mammary gland. However, in women, the molecular mechanisms that underlie this poor prognosis of patients with PP-BCPW remain vastly underexplored, due to of lack of adequate patient numbers and outcome data. EXPERIMENTAL DESIGN We explored whether distinct prognostic features, common to all breast cancer molecular subtypes, exist in postpartum tumor tissue. Using detailed breastfeeding data, we delineated the postweaning period in PP-BC as a surrogate for mammary gland involution and performed whole transcriptome sequencing, immunohistochemical, and (multiplex) immunofluorescent analyses on tumor tissue of patients with PP-BCPW, PP-BCDL, Pr-BC, and NP-BC. RESULTS We found that patients with PP-BCPW having a low expression level of an immunoglobulin gene signature, but high infiltration of plasma B cells, have an increased risk for metastasis and death. Although PP-BCPW tumor tissue was also characterized by an increase in CD8+ cytotoxic T cells and reduced distance among these cell types, these parameters were not associated with differential clinical outcomes among groups. CONCLUSIONS These data point to the importance of plasma B cells in the postweaning mammary tumor microenvironment regarding the poor prognosis of PP-BCPW patients. Future prospective and in-depth research needs to further explore the role of B-cell immunobiology in this specific group of young patients with breast cancer.
Collapse
Affiliation(s)
- Hanne Lefrère
- Department of Oncology, Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
- Department of Gynaecology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kat Moore
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Giuseppe Floris
- Department of Imaging and Pathology, Unit of Translational Cell & Tissue Research, KU Leuven, Leuven, Belgium
- Department of Pathology, Unit of Translational Cell & Tissue Research, University Hospitals Leuven, Leuven, Belgium
- Multidisciplinary Breast Centre, UZ-KU Leuven Cancer Institute (LKI), University Hospitals Leuven, Leuven, Belgium
| | - Joyce Sanders
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Iris M. Seignette
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tycho Bismeijer
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Dennis Peters
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Erik Hooijberg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Kristel Van Calsteren
- Departement of Reproduction and regeneration, Division Women and Child, Feto-Maternal Medicine, KU Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Oncology, Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
- Multidisciplinary Breast Centre, UZ-KU Leuven Cancer Institute (LKI), University Hospitals Leuven, Leuven, Belgium
- Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium
| | - Ellen Warner
- Division of Medical Oncology, Department of Medicine, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Fedro Alessandro Peccatori
- Division of Gynaecological Oncology, Department of Gynaecology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Sibylle Loibl
- German Breast Group, Neu-Isenburg, Hessen, Germany
- Centre for Haematology and Oncology Bethanien, Frankfurt, Germany
| | - Charlotte Maggen
- Department of Oncology, Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Prenatal Medicine, University Hospital Brussels, Brussels, Belgium
| | - Sileny N. Han
- Department of Oncology, Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
- Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium
| | - Katarzyna J. Jerzak
- Division of Medical Oncology, Department of Medicine, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Daniela Annibali
- Department of Oncology, Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Karin E. de Visser
- Oncode Institute, Utrecht, The Netherlands
- Division of Tumour Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lodewyk Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Faculty of EEMCS, Delft University of Technology, Delft, The Netherlands
| | - Liesbeth Lenaerts
- Department of Oncology, Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
| | - Frédéric Amant
- Department of Oncology, Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
- Department of Gynaecology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
10
|
Arora R, Cao C, Kumar M, Sinha S, Chanda A, McNeil R, Samuel D, Arora RK, Matthews TW, Chandarana S, Hart R, Dort JC, Biernaskie J, Neri P, Hyrcza MD, Bose P. Spatial transcriptomics reveals distinct and conserved tumor core and edge architectures that predict survival and targeted therapy response. Nat Commun 2023; 14:5029. [PMID: 37596273 PMCID: PMC10439131 DOI: 10.1038/s41467-023-40271-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 07/19/2023] [Indexed: 08/20/2023] Open
Abstract
The spatial organization of the tumor microenvironment has a profound impact on biology and therapy response. Here, we perform an integrative single-cell and spatial transcriptomic analysis on HPV-negative oral squamous cell carcinoma (OSCC) to comprehensively characterize malignant cells in tumor core (TC) and leading edge (LE) transcriptional architectures. We show that the TC and LE are characterized by unique transcriptional profiles, neighboring cellular compositions, and ligand-receptor interactions. We demonstrate that the gene expression profile associated with the LE is conserved across different cancers while the TC is tissue specific, highlighting common mechanisms underlying tumor progression and invasion. Additionally, we find our LE gene signature is associated with worse clinical outcomes while TC gene signature is associated with improved prognosis across multiple cancer types. Finally, using an in silico modeling approach, we describe spatially-regulated patterns of cell development in OSCC that are predictably associated with drug response. Our work provides pan-cancer insights into TC and LE biology and interactive spatial atlases ( http://www.pboselab.ca/spatial_OSCC/ ; http://www.pboselab.ca/dynamo_OSCC/ ) that can be foundational for developing novel targeted therapies.
Collapse
Affiliation(s)
- Rohit Arora
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christian Cao
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mehul Kumar
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Ayan Chanda
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Reid McNeil
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Divya Samuel
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Rahul K Arora
- Center for Health Informatics, University of Calgary, Calgary, AB, Canada
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - T Wayne Matthews
- Ohlson Research Initiative, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Otolaryngology Head & Neck Surgery, Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shamir Chandarana
- Ohlson Research Initiative, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Otolaryngology Head & Neck Surgery, Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Robert Hart
- Ohlson Research Initiative, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Otolaryngology Head & Neck Surgery, Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Joseph C Dort
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Ohlson Research Initiative, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Otolaryngology Head & Neck Surgery, Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paola Neri
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Division of Hematology, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Martin D Hyrcza
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Pinaki Bose
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
11
|
Fan Y, Wang Y, Wang F, Huang L, Yang Y, Wong KC, Li X. Reliable Identification and Interpretation of Single-Cell Molecular Heterogeneity and Transcriptional Regulation using Dynamic Ensemble Pruning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205442. [PMID: 37290050 PMCID: PMC10401140 DOI: 10.1002/advs.202205442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 05/11/2023] [Indexed: 06/10/2023]
Abstract
Unsupervised clustering is an essential step in identifying cell types from single-cell RNA sequencing (scRNA-seq) data. However, a common issue with unsupervised clustering models is that the optimization direction of the objective function and the final generated clustering labels in the absence of supervised information may be inconsistent or even arbitrary. To address this challenge, a dynamic ensemble pruning framework (DEPF) is proposed to identify and interpret single-cell molecular heterogeneity. In particular, a silhouette coefficient-based indicator is developed to determine the optimization direction of the bi-objective function. In addition, a hierarchical autoencoder is employed to project the high-dimensional data onto multiple low-dimensional latent space sets, and then a clustering ensemble is produced in the latent space by the basic clustering algorithm. Following that, a bi-objective fruit fly optimization algorithm is designed to prune dynamically the low-quality basic clustering in the ensemble. Multiple experiments are conducted on 28 real scRNA-seq datasets and one large real scRNA-seq dataset from diverse platforms and species to validate the effectiveness of the DEPF. In addition, biological interpretability and transcriptional and post-transcriptional regulatory are conducted to explore biological patterns from the cell types identified, which could provide novel insights into characterizing the mechanisms.
Collapse
Affiliation(s)
- Yi Fan
- School of Artificial Intelligence, Jilin University, Jilin, China
| | - Yunhe Wang
- School of Artificial Intelligence, Hebei University of Technology, Tianjin, China
| | - Fuzhou Wang
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong
| | - Lei Huang
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong
| | - Yuning Yang
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong
| | - Xiangtao Li
- School of Artificial Intelligence, Jilin University, Jilin, China
| |
Collapse
|
12
|
Mincarelli L, Uzun V, Wright D, Scoones A, Rushworth SA, Haerty W, Macaulay IC. Single-cell gene and isoform expression analysis reveals signatures of ageing in haematopoietic stem and progenitor cells. Commun Biol 2023; 6:558. [PMID: 37225862 PMCID: PMC10209181 DOI: 10.1038/s42003-023-04936-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
Single-cell approaches have revealed that the haematopoietic hierarchy is a continuum of differentiation, from stem cell to committed progenitor, marked by changes in gene expression. However, many of these approaches neglect isoform-level information and thus do not capture the extent of alternative splicing within the system. Here, we present an integrated short- and long-read single-cell RNA-seq analysis of haematopoietic stem and progenitor cells. We demonstrate that over half of genes detected in standard short-read single-cell analyses are expressed as multiple, often functionally distinct, isoforms, including many transcription factors and key cytokine receptors. We observe global and HSC-specific changes in gene expression with ageing but limited impact of ageing on isoform usage. Integrating single-cell and cell-type-specific isoform landscape in haematopoiesis thus provides a new reference for comprehensive molecular profiling of heterogeneous tissues, as well as novel insights into transcriptional complexity, cell-type-specific splicing events and consequences of ageing.
Collapse
Affiliation(s)
- Laura Mincarelli
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, United Kingdom.
| | - Vladimir Uzun
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, United Kingdom
| | - David Wright
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, United Kingdom
| | - Anita Scoones
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, United Kingdom
| | - Stuart A Rushworth
- Norwich Medical School, The University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Wilfried Haerty
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, United Kingdom.
| | - Iain C Macaulay
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, United Kingdom.
| |
Collapse
|
13
|
Milardi G, Lleo A. Tumor-Infiltrating B Lymphocytes: Promising Immunotherapeutic Targets for Primary Liver Cancer Treatment. Cancers (Basel) 2023; 15:2182. [PMID: 37046842 PMCID: PMC10093314 DOI: 10.3390/cancers15072182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Hepatocellular carcinoma and cholangiocarcinoma are the fourth most lethal primary cancers worldwide. Therefore, there is an urgent need for therapeutic strategies, including immune cell targeting therapies. The heterogeneity of liver cancer is partially explained by the characteristics of the tumor microenvironment (TME), where adaptive and innate immune system cells are the main components. Pioneering studies of primary liver cancers revealed that tumor-infiltrating immune cells and their dynamic interaction with cancer cells significantly impacted carcinogenesis, playing an important role in cancer immune evasion and responses to immunotherapy treatment. In particular, B cells may play a prominent role and have a controversial function in the TME. In this work, we highlight the effect of B lymphocytes as tumor infiltrates in relation to primary liver cancers and their potential prognostic value. We also present the key pathways underlying B-cell interactions within the TME, as well as the way that a comprehensive characterization of B-cell biology can be exploited to develop novel immune-based therapeutic approaches.
Collapse
Affiliation(s)
- Giulia Milardi
- Hepatobiliary Immunopathology Labaratory, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
- Department of Gastroenterology, Division of Internal Medicine and Hepatology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| |
Collapse
|
14
|
Chandler KB, Pavan CH, Cotto Aparicio HG, Sackstein R. Enrichment and nLC-MS/MS Analysis of Head and Neck Cancer Mucinome Glycoproteins. J Proteome Res 2023; 22:1231-1244. [PMID: 36971183 DOI: 10.1021/acs.jproteome.2c00746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Mucin-domain glycoproteins expressed on cancer cell surfaces play central roles in cell adhesion, cancer progression, stem cell renewal, and immune evasion. Despite abundant evidence that mucin-domain glycoproteins are critical to the pathobiology of head and neck squamous cell carcinoma (HNSCC), our knowledge of the composition of that mucinome is grossly incomplete. Here, we utilized a catalytically inactive point mutant of the enzyme StcE (StcEE447D) to capture mucin-domain glycoproteins in head and neck cancer cell line lysates followed by their characterization using sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE), in-gel digestion, nano-liquid chromatography-tandem mass spectrometry (nLC-MS/MS), and enrichment analyses. We demonstrate the feasibility of this workflow for the study of mucin-domain glycoproteins in HNSCC, identify a set of mucin-domain glycoproteins common to multiple HNSCC cell lines, and report a subset of mucin-domain glycoproteins that are uniquely expressed in HSC-3 cells, a cell line derived from a highly aggressive metastatic tongue squamous cell carcinoma. This effort represents the first attempt to identify mucin-domain glycoproteins in HNSCC in an untargeted, unbiased analysis, paving the way for a more comprehensive characterization of the mucinome components that mediate aggressive tumor cell phenotypes. Data associated with this study have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD029420.
Collapse
|
15
|
Ma H, Murphy C, Loscher CE, O’Kennedy R. Autoantibodies - enemies, and/or potential allies? Front Immunol 2022; 13:953726. [PMID: 36341384 PMCID: PMC9627499 DOI: 10.3389/fimmu.2022.953726] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/24/2022] [Indexed: 08/13/2023] Open
Abstract
Autoantibodies are well known as potentially highly harmful antibodies which attack the host via binding to self-antigens, thus causing severe associated diseases and symptoms (e.g. autoimmune diseases). However, detection of autoantibodies to a range of disease-associated antigens has enabled their successful usage as important tools in disease diagnosis, prognosis and treatment. There are several advantages of using such autoantibodies. These include the capacity to measure their presence very early in disease development, their stability, which is often much better than their related antigen, and the capacity to use an array of such autoantibodies for enhanced diagnostics and to better predict prognosis. They may also possess capacity for utilization in therapy, in vivo. In this review both the positive and negative aspects of autoantibodies are critically assessed, including their role in autoimmune diseases, cancers and the global pandemic caused by COVID-19. Important issues related to their detection are also highlighted.
Collapse
Affiliation(s)
- Hui Ma
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Caroline Murphy
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | - Richard O’Kennedy
- School of Biotechnology, Dublin City University, Dublin, Ireland
- Research, Development and Innovation, Qatar Foundation, Doha, Qatar
- Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
16
|
B-Cell-Based Immunotherapy: A Promising New Alternative. Vaccines (Basel) 2022; 10:vaccines10060879. [PMID: 35746487 PMCID: PMC9227543 DOI: 10.3390/vaccines10060879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
The field of immunotherapy has undergone radical conceptual changes over the last decade. There are various examples of immunotherapy, including the use of monoclonal antibodies, cancer vaccines, tumor-infecting viruses, cytokines, adjuvants, and autologous T cells carrying chimeric antigen receptors (CARs) that can bind cancer-specific antigens known as adoptive immunotherapy. While a lot has been achieved in the field of T-cell immunotherapy, only a fraction of patients (20%) see lasting benefits from this mode of treatment, which is why there is a critical need to turn our attention to other immune cells. B cells have been shown to play both anti- and pro-tumorigenic roles in tumor tissue. In this review, we shed light on the dual nature of B cells in the tumor microenvironment. Furthermore, we discussed the different factors affecting the biology and function of B cells in tumors. In the third section, we described B-cell-based immunotherapies and their clinical applications and challenges. These current studies provide a springboard for carrying out future mechanistic studies to help us unleash the full potential of B cells in immunotherapy.
Collapse
|
17
|
Aziz S, Rasheed F, Zahra R, König S. Gastric Cancer Pre-Stage Detection and Early Diagnosis of Gastritis Using Serum Protein Signatures. Molecules 2022; 27:molecules27092857. [PMID: 35566209 PMCID: PMC9099457 DOI: 10.3390/molecules27092857] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
Background: A gastric cancer (GC) diagnosis relies on histopathology. Endoscopy rates are increasing. Helicobacter pylori infection is a major GC risk factor. In an effort to elucidate abundant blood biomarkers, and potentially reduce the number of diagnostic surgical interventions, we investigated sera and biopsies from a cohort of 219 H. pylori positive and negative patients diagnosed with GC, gastritis, and ulcers. This allowed the comparative investigation of the different gastroduodenal diseases, and the exclusion of protein changes resulting from bacterial infection or inflammation of the gastric mucosa when searching for GC-dependent proteins. Methods: High-definition mass spectrometry-based expression analysis of tryptically digested proteins was performed, followed by multivariate statistical and network analyses for the different disease groups, with respect to H. pylori infection status. Significantly regulated proteins differing more than two-fold between groups were shortlisted, and their role in gastritis and GC discussed. Results: We present data of comparative protein analyses of biopsies and sera from patients suffering from mild to advanced gastritis, ulcers, and early to advanced GC, in conjunction with a wealth of metadata, clinical information, histopathological evaluation, and H. pylori infection status. We used samples from pre-malignant stages to extract prospective serum markers for early-stage GC, and present a 29-protein marker panel containing, amongst others, integrin β-6 and glutathione peroxidase. Furthermore, ten serum markers specific for advanced GC, independent of H. pylori infection, are provided. They include CRP, protein S100A9, and kallistatin. The majority of these proteins were previously discussed in the context of cancer or GC. In addition, we detected hypoalbuminemia and increased fibrinogen serum levels in gastritis. Conclusion: Two protein panels were suggested for the development of multiplex tests for GC serum diagnostics. For most of the elements contained in these panels, individual commercial tests are available. Thus, we envision the design of multi-protein assays, incorporating several to all of the panel members, in order to gain a level of specificity that cannot be achieved by testing a single protein alone. As their development and validation will take time, gastritis diagnosis based on the fibrinogen to albumin serum ratio may be a quick way forward. Its determination at the primary/secondary care level for early diagnosis could significantly reduce the number of referrals to endoscopy. Preventive measures are in high demand. The protein marker panels presented in this work will contribute to improved GC diagnostics, once they have been transferred from a research result to a practical tool.
Collapse
Affiliation(s)
- Shahid Aziz
- BreathMAT Lab, Pakistan Institute of Nuclear Science and Technology (PINSTEC), Islamabad 44000, Pakistan; (S.A.); (F.R.)
- Department of Microbiology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan;
- IZKF Core Unit Proteomics, University of Münster, 48149 Münster, Germany
| | - Faisal Rasheed
- BreathMAT Lab, Pakistan Institute of Nuclear Science and Technology (PINSTEC), Islamabad 44000, Pakistan; (S.A.); (F.R.)
| | - Rabaab Zahra
- Department of Microbiology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Simone König
- IZKF Core Unit Proteomics, University of Münster, 48149 Münster, Germany
- Correspondence:
| |
Collapse
|