1
|
Cannizzaro IR, Treccani M, Taiani A, Ambrosini E, Busciglio S, Cesarini S, Luberto A, De Sensi E, Moschella B, Gismondi P, Azzoni C, Bottarelli L, Giordano G, Corradi D, Silini EM, Zanatta V, Cennamo F, Bertolini P, Caggiati P, Martorana D, Uliana V, Percesepe A, Barili V. Proof of Concept for Genome Profiling of the Neurofibroma/Sarcoma Sequence in Neurofibromatosis Type 1. Int J Mol Sci 2024; 25:10822. [PMID: 39409151 PMCID: PMC11476461 DOI: 10.3390/ijms251910822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/27/2024] [Accepted: 10/06/2024] [Indexed: 10/20/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disorder characterized by the predisposition to develop tumors such as malignant peripheral nerve sheath tumors (MPNSTs) which represents the primary cause of death for NF1-affected patients. Regardless of the high incidence and mortality, the molecular mechanisms underneath MPNST growth and metastatic progression remain poorly understood. In this proof-of-concept study, we performed somatic whole-exome sequencing (WES) to profile the genomic alterations in four samples from a patient with NF1-associated MPNST, consisting of a benign plexiform neurofibroma, a primary MPNST, and metastases from lung and skin tissues. By comparing genomic patterns, we identified a high level of variability across samples with distinctive genetic changes which allow for the definition of profiles of the early phase with respect to the late metastatic stages. Pathogenic and likely pathogenic variants were abundant in the primary tumor, whereas the metastatic samples exhibited a high level of copy-number variations (CNVs), highlighting a possible genomic instability in the late phases. The most known MPNST-related genes, such as TP53 and SUZ12, were identified in CNVs observed within the primary tumor. Pathway analysis of altered early genes in MPNST pointed to a potential role in cell motility, division and metabolism. Moreover, we employed survival analysis with the TCGA sarcoma genomic dataset on 262 affected patients, in order to corroborate the predictive significance of the identified early and metastatic MPNST driver genes. Specifically, the expression changes related to the mutated genes, such as in RBMX, PNPLA6 and AGAP2, were associated with reduced patient survival, distinguishing them as potential prognostic biomarkers. This study underlines the relevance of integrating genomic results with clinical information for early diagnosis and prognostic understanding of tumor aggressiveness.
Collapse
Affiliation(s)
- Ilenia Rita Cannizzaro
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Mirko Treccani
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Human Nutrition Unit, Department of Food and Drug, University of Parma, 43125 Parma, Italy
| | - Antonietta Taiani
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Enrico Ambrosini
- Medical Genetics, University Hospital of Parma, 43126 Parma, Italy
| | - Sabrina Busciglio
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Sofia Cesarini
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Anita Luberto
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Erika De Sensi
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Barbara Moschella
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Pierpacifico Gismondi
- Pediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Cinzia Azzoni
- Pathology Unit, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Lorena Bottarelli
- Pathology Unit, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Giovanna Giordano
- Pathology Unit, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Domenico Corradi
- Pathology Unit, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Enrico Maria Silini
- Pathology Unit, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Valentina Zanatta
- Cytogenetics, Molecular Genetics and Medical Genetics Unit, Toma Advanced Biomedical Assays, 21052 Busto Arsizio, Italy
| | - Federica Cennamo
- Pediatric Hematology Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Patrizia Bertolini
- Pediatric Hematology Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | | | - Davide Martorana
- Medical Genetics, University Hospital of Parma, 43126 Parma, Italy
| | - Vera Uliana
- Medical Genetics, University Hospital of Parma, 43126 Parma, Italy
| | - Antonio Percesepe
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Medical Genetics, University Hospital of Parma, 43126 Parma, Italy
| | - Valeria Barili
- Medical Genetics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
2
|
Napoli M, Bauer J, Bonod C, Goff SVL, Moali C. PCPE-2 (procollagen C-proteinase enhancer-2): the NON-IDENTICAL twin of PCPE-1. Matrix Biol 2024:S0945-053X(24)00113-6. [PMID: 39251075 DOI: 10.1016/j.matbio.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
PCPE-2 was discovered at the beginning of this century, and was soon identified as a close homolog of PCPE-1 (procollagen C-proteinase enhancer 1). After the demonstration that it could also stimulate the proteolytic maturation of fibrillar procollagens by BMP-1/tolloid-like proteinases (BTPs), PCPE-2 did not attract much attention as it was thought to fulfill the same functions as PCPE-1 which was already well-described. However, the tissue distribution of PCPE-2 shows both common points and significant differences with PCPE-1, suggesting that their activities are not fully overlapping. Also, the recently established connections between PCPE-2 (gene name PCOLCE2) and several important diseases such as atherosclerosis, inflammatory diseases and cancer have highlighted the need for a thorough reappraisal of the in vivo roles of this regulatory protein. In this context, the recent finding that, while retaining the ability to bind fibrillar procollagens and to activate their C-terminal maturation, PCPE-2 can also bind BTPs and inhibit their activity has substantially extended its potential functions. In this review, we describe the current knowledge about PCPE-2 with a focus on collagen fibrillogenesis, lipid metabolism and inflammation, and discuss how we could further advance our understanding of PCPE-2-dependent biological processes.
Collapse
Affiliation(s)
- Manon Napoli
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Julien Bauer
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Christelle Bonod
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Sandrine Vadon-Le Goff
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Catherine Moali
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France.
| |
Collapse
|
3
|
Hong J, Jin HJ, Choi MR, Lim DWT, Park JE, Kim YS, Lim SB. Matrisomics: Beyond the extracellular matrix for unveiling tumor microenvironment. Biochim Biophys Acta Rev Cancer 2024; 1879:189178. [PMID: 39241895 DOI: 10.1016/j.bbcan.2024.189178] [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: 06/05/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
The matrisome, a group of proteins constituting or interacting with the extracellular matrix (ECM), has garnered attention as a potent regulator of cancer progression. An increasing number of studies have focused on cancer matrisome utilizing diverse -omics approaches. Here, we present diverse patterns of matrisomal populations within cancer tissues, exploring recent -omics studies spanning different '-omics' levels (epigenomics, genomics, transcriptomics, and proteomics), as well as newly developed sequencing techniques such as single-cell RNA sequencing and spatial transcriptomics. Some matrisome genes showed uniform patterns of upregulated or downregulated expression across various cancers, while others displayed different expression patterns according to the cancer types. This matrisomal dysregulation in cancer was further examined according to their originating cell type and spatial location in the tumor tissue. Experimental studies were also collected to demonstrate the identified roles of matrisome genes during cancer progression. Interestingly, many studies on cancer matrisome have suggested matrisome genes as effective biomarkers in cancer research. Although the specific mechanisms and clinical applications of cancer matrisome have not yet been fully elucidated, recent techniques and analyses on cancer matrisomics have emphasized their biological importance in cancer progression and their clinical implications in deciding the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Jiwon Hong
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Hyo Joon Jin
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Mi Ran Choi
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Darren Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre, Singapore 168583, Singapore
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - You-Sun Kim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
4
|
Yue L, Gong T, Jiang W, Qian L, Gong W, Sun Y, Cai X, Xu H, Liu F, Wang H, Li S, Zhu Y, Zheng Z, Wu Q, Guo T. Proteomic profiling of ovarian clear cell carcinomas identifies prognostic biomarkers for chemotherapy. Proteomics 2024; 24:e2300242. [PMID: 38171885 DOI: 10.1002/pmic.202300242] [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: 06/08/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Clear cell ovarian carcinoma (CCOC) is a relatively rare subtype of ovarian cancer (OC) with high degree of resistance to standard chemotherapy. Little is known about the underlying molecular mechanisms, and it remains a challenge to predict its prognosis after chemotherapy. Here, we first analyzed the proteome of 35 formalin-fixed paraffin-embedded (FFPE) CCOC tissue specimens from a cohort of 32 patients with CCOC (H1 cohort) and characterized 8697 proteins using data-independent acquisition mass spectrometry (DIA-MS). We then performed proteomic analysis of 28 fresh frozen (FF) CCOC tissue specimens from an independent cohort of 24 patients with CCOC (H2 cohort), leading to the identification of 9409 proteins with DIA-MS. After bioinformatics analysis, we narrowed our focus to 15 proteins significantly correlated with the recurrence free survival (RFS) in both cohorts. These proteins are mainly involved in DNA damage response, extracellular matrix (ECM), and mitochondrial metabolism. Parallel reaction monitoring (PRM)-MS was adopted to validate the prognostic potential of the 15 proteins in the H1 cohort and an independent confirmation cohort (H3 cohort). Interferon-inducible transmembrane protein 1 (IFITM1) was observed as a robust prognostic marker for CCOC in both PRM data and immunohistochemistry (IHC) data. Taken together, this study presents a CCOC proteomic data resource and a single promising protein, IFITM1, which could potentially predict the recurrence and survival of CCOC.
Collapse
Affiliation(s)
- Liang Yue
- School of Life Sciences, Fudan University, Shanghai, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Tingting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University
| | - Wenhao Jiang
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Liujia Qian
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Wangang Gong
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yaoting Sun
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Xue Cai
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Heli Xu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fanghua Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Wang
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Sainan Li
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- National Health Commission Key Laboratory of Reproductive Health, Institute of Reproductive and Child Health, Peking University, Beijing, China
| | - Yi Zhu
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Zhiguo Zheng
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Qijun Wu
- Department of Clinical Epidemiology, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tiannan Guo
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Padathpeedika Khalid J, Mary Martin T, Prathap L, Abhimanyu Nisargandha M, Boopathy N, Kishore Kumar MS. Exploring Tumor-Promoting Qualities of Cancer-Associated Fibroblasts and Innovative Drug Discovery Strategies With Emphasis on Thymoquinone. Cureus 2024; 16:e53949. [PMID: 38468988 PMCID: PMC10925941 DOI: 10.7759/cureus.53949] [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: 12/13/2023] [Accepted: 02/09/2024] [Indexed: 03/13/2024] Open
Abstract
Tumor epithelial development and chemoresistance are highly promoted by the tumor microenvironment (TME), which is mostly made up of the cancer stroma. This is due to several causes. Cancer-associated fibroblasts (CAFs) stand out among them as being essential for the promotion of tumors. Understanding the fibroblastic population within a single tumor is made more challenging by the undeniable heterogeneity within it, even though particular stromal alterations are still up for debate. Numerous chemical signals released by tumors improve the connections between heterotypic fibroblasts and CAFs, promoting the spread of cancer. It becomes essential to have a thorough understanding of this complex microenvironment to effectively prevent solid tumor growth. Important new insights into the role of CAFs in the TME have been revealed by recent studies. The objective of this review is to carefully investigate the relationship between CAFs in tumors and plant secondary metabolites, with a focus on thymoquinone (TQ). The literature published between 2010 and 2023 was searched in PubMed and Google Scholar with keywords such as TQ, TME, cancer-associated fibroblasts, mechanism of action, and flavonoids. The results showed a wealth of data substantiating the activity of plant secondary metabolites, particularly TQ's involvement in blocking CAF operations. Scrutinized research also clarified the wider effect of flavonoids on pathways related to cancer. The present study highlights the complex dynamics of the TME and emphasizes the critical role of CAFs. It also examines the possible interventions provided by secondary metabolites found in plants, with TQ playing a vital role in regulating CAF function based on recent literature.
Collapse
Affiliation(s)
- Jabir Padathpeedika Khalid
- Department of Physiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Taniya Mary Martin
- Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Lavanya Prathap
- Department of Anatomy, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Milind Abhimanyu Nisargandha
- Department of Physiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Nisha Boopathy
- Department of Community Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Meenakshi Sundaram Kishore Kumar
- Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
6
|
Ulaganathan K, Puranam K, Mukta S, Hanumanth SR. Expression profiling of luminal B breast tumor in Indian women. J Cancer Res Clin Oncol 2023; 149:13645-13664. [PMID: 37516983 DOI: 10.1007/s00432-023-05195-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 08/01/2023]
Abstract
PURPOSE In this study, we aimed at profiling of luminal B breast cancer specific gene expression pattern in Indian women using mRNA-seq and validation based on TCGA expression data. METHODS RNA isolated from luminal B tumor and adjacent normal tissues was used for library construction and sequencing. Reference-based assemblies of these reads were used for differential gene expression analysis using DeSeq2. The DEGs were evaluated using TCGA expression data. Kaplan-Meier survival method was used to evaluate association between genes showing luminal B specific differential expression pattern and breast cancer prognosis and statistical significance was assessed using log-rank test. Alternate splicing analysis was done using rmats. RESULTS Differential expression analysis identified 2371 differentially expressed genes (DEGs) in luminal B breast tumors in comparison with adjacent normal tissues of Indian Women. Of them, 1692 DEGs were validated using TCGA luminal B paired samples. Integration of this data with the DEGs obtained by comparative analysis of unpaired luminal B with luminal A unpaired samples from TCGA resulted in 291 DEGs showing luminal B specific expression pattern. Further, 26 genes of prognostic value were identified. Differential splicing analysis between luminal B tumors and adjacent normal tissues in our cohort led to the identification of 687 genes showing significant differential alternate splicing events. CONCLUSION This study profiled gene expression pattern of luminal B tumors of Indian women and identified 26 key genes of prognostic value for luminal B breast cancer. This study also profiled differential alternate splicing and identified important alternate splicing events in luminal B breast cancer.
Collapse
Affiliation(s)
| | - Kaushik Puranam
- Department of Genetics, Osmania University, Hyderabad, Telangana, 500007, India
| | - Srinivasulu Mukta
- Department of Surgical Oncology, MNJ Institute of Oncology and RCC, Hyderabad, Telangana, India
| | | |
Collapse
|
7
|
Yu D, Yang P, Lu X, Huang S, Liu L, Fan X. Single-cell RNA sequencing reveals enhanced antitumor immunity after combined application of PD-1 inhibitor and Shenmai injection in non-small cell lung cancer. Cell Commun Signal 2023; 21:169. [PMID: 37430270 DOI: 10.1186/s12964-023-01184-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/04/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have altered the clinical management of non-small cell lung cancer (NSCLC). However, the low response rate, severe immune-related adverse events (irAEs), and hyperprogressive disease following ICIs monotherapy require attention. Combination therapy may overcome these limitations and traditional Chinese medicine with immunomodulatory effects provides a promising approach. Shenmai injection (SMI) is a clinically effective adjuvant treatment for cancer with chemotherapy and radiotherapy. Therefore, the combined effects and mechanisms of SMI and programmed death-1 (PD-1) inhibitor against NSCLC was focused on this study. METHODS A Lewis lung carcinoma mouse model and a lung squamous cell carcinoma humanized mouse model were used to investigate the combined efficacy and safety of SMI and PD-1 inhibitor. The synergistic mechanisms of the combination therapy against NSCLC were explored using single-cell RNA sequencing. Validation experiments were performed using immunofluorescence analysis, in vitro experiment, and bulk transcriptomic datasets. RESULTS In both models, combination therapy alleviated tumor growth and prolonged survival without increasing irAEs. The GZMAhigh and XCL1high natural killer (NK) cell subclusters with cytotoxic and chemokine signatures increased in the combination therapy, while malignant cells from combination therapy were mainly in the apoptotic state, suggesting that mediating tumor cell apoptosis through NK cells is the main synergistic mechanisms of combination therapy. In vitro experiment confirmed that combination therapy increased secretion of Granzyme A by NK cells. Moreover, we discovered that PD-1 inhibitor and SMI combination blocked inhibitory receptors on NK and T cells and restores their antitumoral activity in NSCLC better than PD-1 inhibitor monotherapy, and immune and stromal cells exhibited a decrease of angiogenic features and attenuated cancer metabolism reprogramming in microenvironment of combination therapy. CONCLUSIONS This study demonstrated that SMI reprograms tumor immune microenvironment mainly by inducing NK cells infiltration and synergizes with PD-1 inhibitor against NSCLC, suggested that targeting NK cells may be an important strategy for combining with ICIs. Video Abstract.
Collapse
Affiliation(s)
- Dingyi Yu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Penghui Yang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Shaoze Huang
- Zhejiang Engineering Research Center for Advanced Manufacturing of Traditional Chinese Medicine, Huzhou, China
| | - Li Liu
- Zhejiang Engineering Research Center for Advanced Manufacturing of Traditional Chinese Medicine, Huzhou, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| |
Collapse
|
8
|
Okuda-Ashitaka E, Matsumoto KI. Tenascin-X as a causal gene for classical-like Ehlers-Danlos syndrome. Front Genet 2023; 14:1107787. [PMID: 37007968 PMCID: PMC10050494 DOI: 10.3389/fgene.2023.1107787] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Tenascin-X (TNX) is an extracellular matrix glycoprotein for which a deficiency results in a recessive form of classical-like Ehlers-Danlos syndrome (clEDS), a heritable connective tissue disorder with hyperextensible skin without atrophic scarring, joint hypermobility, and easy bruising. Notably, patients with clEDS also suffer from not only chronic joint pain and chronic myalgia but also neurological abnormalities such as peripheral paresthesia and axonal polyneuropathy with high frequency. By using TNX-deficient (Tnxb−/−) mice, well-known as a model animal of clEDS, we recently showed that Tnxb−/− mice exhibit hypersensitivity to chemical stimuli and the development of mechanical allodynia due to the hypersensitization of myelinated A-fibers and activation of the spinal dorsal horn. Pain also occurs in other types of EDS. First, we review the underlying molecular mechanisms of pain in EDS, especially that in clEDS. In addition, the roles of TNX as a tumor suppressor protein in cancer progression have been reported. Recent in silico large-scale database analyses have shown that TNX is downregulated in various tumor tissues and that high expression of TNX in tumor cells has a good prognosis. We describe what is so far known about TNX as a tumor suppressor protein. Furthermore, some patients with clEDS show delayed wound healing. Tnxb−/− mice also exhibit impairment of epithelial wound healing in corneas. TNX is also involved in liver fibrosis. We address the molecular mechanism for the induction of COL1A1 by the expression of both a peptide derived from the fibrinogen-related domain of TNX and integrin α11.
Collapse
Affiliation(s)
- Emiko Okuda-Ashitaka
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka, Japan
- *Correspondence: Emiko Okuda-Ashitaka, ; Ken-ichi Matsumoto,
| | - Ken-ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Head Office for Research and Academic Information, Shimane University, Izumo, Japan
- *Correspondence: Emiko Okuda-Ashitaka, ; Ken-ichi Matsumoto,
| |
Collapse
|
9
|
Wang F, Yan X, Hua Y, Song J, Liu D, Yang C, Peng F, Kang F, Hui Y. PI3K/AKT/mTOR pathway and its related molecules participate in PROK1 silence-induced anti-tumor effects on pancreatic cancer. Open Life Sci 2023; 18:20220538. [PMID: 37070074 PMCID: PMC10105552 DOI: 10.1515/biol-2022-0538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/28/2022] [Accepted: 11/21/2022] [Indexed: 04/19/2023] Open
Abstract
The PI3K/AKT/mTOR (phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin) pathway can be initiated by PROK1 (prokineticin 1), but its effect and mechanism of action in pancreatic carcinoma (PC) are not fully understood. In this study, we elucidated the roles of PROK1 and its related molecules in PC in vivo. PANC-1 cells with PROK1 knockdown were injected into BALB/c nude mice. The growth and weight of the tumor were monitored and measured, which was followed by TUNEL (terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling), immunohistochemical staining, and hematoxylin and eosin staining. The key proteins related to proliferation, apoptosis, and the PI3K/AKT/mTOR pathway were determined by Western blotting. We also used public databases to identify the molecules related to PROK1. The reduction of PROK1 inhibited angiopoiesis and promoted apoptosis in vivo. PCNA-1, cyclin D1, and Bcl-2 decreased considerably, while Bax and cleaved caspase-3 increased significantly after PROK1 inhibition. The PI3K/AKT/mTOR signal inhibition was also closely associated with PROK1 knockdown. The possible related molecules of PROK1, such as von Willebrand factor, were screened and considered to be involved in the aberrant activation of PI3K/AKT. In conclusion, PROK1 knockdown significantly prevented tumor growth and promoted apoptosis of human PC cells in vivo, where the PI3K/AKT/mTOR pathway was probably inhibited. Therefore, PROK1, along with its related molecules, might be important targets for PC therapy.
Collapse
Affiliation(s)
- Feng Wang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Xiaogang Yan
- Department of Surgical Oncology, The First People’s Hospital of Yinchuan, Yinchuan750001, China
| | - Yongqiang Hua
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, Shanghai200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Jianjun Song
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Di Liu
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Chun Yang
- Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan750001, China
| | - Fei Peng
- Department of Hepatobiliary Pancreatic Surgery, Edong Healthcare Huangshi Central Hospital, Huangshi435002, Hubei, China
| | - Fuping Kang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Yongfeng Hui
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| |
Collapse
|
10
|
Zhu W, Feng D, Shi X, Li D, Wei Q, Yang L. A pan-cancer analysis of the oncogenic role of zinc finger protein 419 in human cancer. Front Oncol 2022; 12:1042118. [PMID: 36578929 PMCID: PMC9791222 DOI: 10.3389/fonc.2022.1042118] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background As a ferroptosis-related gene, the polymorphism of zinc finger protein 419 (ZNF419) at the splice donor site may generate renal cell carcinoma-associated novel minor histocompatibility antigen ZAPHIR. However, the role of ZNF419 in prognosis and immunology in human tumors remains largely unknown. This study aimed to visualize the prognostic landscape of ZNF419 at pan-cancer level and explore the relationship between ZNF419 expression and the tumor immune microenvironment. Method Pan-cancer and mutation data were downloaded from TCGA databases and analyzed through R (version 3.6.4) and its suitable packages. Differential ZNF419 expression and prognosis were analyzed. Correlations with ferroptosis-related genes, pathway analysis, tumor stemness, heterogeneity, mutation landscape, and RNA modifications were also explored. The relationships between ZNF419 expression and tumor immunity were investigated through the TIMER and ESTIMATE methods. Result ZNF419 was differentially expressed between tumor and normal samples and was associated with overall survival, disease-specific survival and progression-free interval for STES, KIRC, LIHC, LUSC, PRAD, and BLCA. We found the interaction between ZNF419 and FANCD2 might involve in ferroptosis in pan-cancer level. In addition, the mutation frequencies of STES, KIRC, LIHC, LUSC, PRAD, and BLCA were 1.5%, 0.3%, 0.3%, 1.9%, 0.2%, and 0.7%, respectively. We detected that the expression of ZNF419 was closely correlated with most immune checkpoint genes and immune regulatory genes. Furthermore, we found that the ZNF419 expression level was negatively related to the immune score in the six cancers mentioned above. The expression of ZNF419 was significantly associated with various infiltrating immune cells, such as CD4+ T cells, CD8+ T cells, and macrophages in patients with KIRC, PRAD, and LUSC but was only significantly related to macrophages in BLCA patients. Conclusion ZNF419 might serve as a potential prognostic and immunological pan-cancer biomarker, especially for KIRC, LIHC, LUSC, PRAD, and BLCA.
Collapse
Affiliation(s)
| | | | | | | | - Qiang Wei
- *Correspondence: Qiang Wei, ; Lu Yang,
| | - Lu Yang
- *Correspondence: Qiang Wei, ; Lu Yang,
| |
Collapse
|
11
|
Urh K, Zidar N, Boštjančič E. Bioinformatics Analysis of RNA-seq Data Reveals Genes Related to Cancer Stem Cells in Colorectal Cancerogenesis. Int J Mol Sci 2022; 23:ijms232113252. [PMID: 36362041 PMCID: PMC9654446 DOI: 10.3390/ijms232113252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Cancer stem cells (CSC) play one of the crucial roles in the pathogenesis of various cancers, including colorectal cancer (CRC). Although great efforts have been made regarding our understanding of the cancerogenesis of CRC, CSC involvement in CRC development is still poorly understood. Using bioinformatics and RNA-seq data of normal mucosa, colorectal adenoma, and carcinoma (n = 106) from GEO and TCGA, we identified candidate CSC genes and analyzed pathway enrichment analysis (PEI) and protein–protein interaction analysis (PPI). Identified CSC-related genes were validated using qPCR and tissue samples from 47 patients with adenoma, adenoma with early carcinoma, and carcinoma without and with lymph node metastasis and were compared to normal mucosa. Six CSC-related genes were identified: ANLN, CDK1, ECT2, PDGFD, TNC, and TNXB. ANLN, CDK1, ECT2, and TNC were differentially expressed between adenoma and adenoma with early carcinoma. TNC was differentially expressed in CRC without lymph node metastases whereas ANLN, CDK1, and PDGFD were differentially expressed in CRC with lymph node metastases compared to normal mucosa. ANLN and PDGFD were differentially expressed between carcinoma without and with lymph node metastasis. Our study identified and validated CSC-related genes that might be involved in early stages of CRC development (ANLN, CDK1, ECT2, TNC) and in development of metastasis (ANLN, PDGFD).
Collapse
|
12
|
Barrón-Gallardo CA, Garcia-Chagollán M, Morán-Mendoza AJ, Delgadillo-Cristerna R, Martínez-Silva MG, Villaseñor-García MM, Aguilar-Lemarroy A, Jave-Suárez LF. A gene expression signature in HER2+ breast cancer patients related to neoadjuvant chemotherapy resistance, overall survival, and disease-free survival. Front Genet 2022; 13:991706. [PMID: 36338974 PMCID: PMC9634254 DOI: 10.3389/fgene.2022.991706] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
Breast cancer ranks first in terms of mortality and incidence rates worldwide among women. The HER2+ molecular subtype is one of the most aggressive subtypes; its treatment includes neoadjuvant chemotherapy and the use of a HER2 antibody. Some patients develop resistance despite positive results obtained using this therapeutic strategy. Objective. To identify prognostic markers for treatment and survival in HER2+ patients. Methods. Patients treated with neoadjuvant chemotherapy were assigned to sensitive and resistant groups based on their treatment response. Differentially expressed genes (DEGs) were identified using RNA-seq analysis. KEGG pathway, gene ontology, and interactome analyses were performed for all DEGs. An enrichment analysis Gene set enrichment analysis was performed. All DEGs were analyzed for overall (OS) and disease-free survival (DFS). Results. A total of 94 DEGs were related to treatment resistance. Survival analysis showed that 12 genes (ATF6B, DHRS13, DIRAS1, ERAL1, GRIN2B, L1CAM, IRX3, PRTFDC1, PBX2, S100B, SLC9A3R2, and TNXB) were good predictors of disease-free survival, and eight genes (GNG4, IL22RA2, MICA, S100B, SERPINF2, HLA-A, DIRAS1, and TNXB) were good predictors of overall survival (OS). Conclusion: We highlighted a molecular expression signature that can differentiate the treatment response, overall survival, and DFS of patients with HER2+ breast cancer.
Collapse
Affiliation(s)
- Carlos A. Barrón-Gallardo
- Programa de Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Mariel Garcia-Chagollán
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Andres J. Morán-Mendoza
- Hospital de Ginecología, Centro Médico Nacional de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
| | - Raul Delgadillo-Cristerna
- Departamento de Radiología e Imagen, Centro Médico Nacional de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
| | - María G. Martínez-Silva
- Departamento de Anatomía Patológica, Centro Médico Nacional de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
| | - María M. Villaseñor-García
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
| | - Adriana Aguilar-Lemarroy
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
- *Correspondence: Adriana Aguilar-Lemarroy, ; Luis F. Jave-Suárez,
| | - Luis F. Jave-Suárez
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara, Mexico
- *Correspondence: Adriana Aguilar-Lemarroy, ; Luis F. Jave-Suárez,
| |
Collapse
|
13
|
A pan-cancer analysis of matrisome proteins reveals CTHRC1 and a related network as major ECM regulators across cancers. PLoS One 2022; 17:e0270063. [PMID: 36190948 PMCID: PMC9529084 DOI: 10.1371/journal.pone.0270063] [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: 02/04/2022] [Accepted: 06/02/2022] [Indexed: 11/07/2022] Open
Abstract
The extracellular matrix in the tumour microenvironment can regulate cancer cell growth and progression. A pan-cancer analysis of TCGA data from 30 cancer types, identified the top 5% of matrisome genes with amplifications or deletions in their copy number, that affect their expression and cancer survival. A similar analysis of matrisome genes in individual cancers identified CTHRC1 to be significantly altered. CTHRC1, a regulator of collagen synthesis, was identified as the most prominently upregulated matrisome gene of interest across cancers. Differential gene expression analysis identified 19 genes whose expression is increased with CTHRC1. STRING analysis of these genes classified them as ‘extracellular’, involved most prominently in ECM organization and cell adhesion. KEGG analysis showed their involvement in ECM-receptor and growth factor signalling. Cytohubba analysis of these genes revealed 13 hub genes, of which MMP13, POSTN, SFRP4, ADAMTS16 and FNDC1 were significantly altered in their expression with CTHRC1 and seen to affect survival across cancers. This could in part be mediated by their overlapping roles in regulating ECM (collagen or fibronectin) expression and organisation. In breast cancer tumour samples CTHRC1 protein levels are significantly upregulated with POSTN and MMP13, further supporting the need to evaluate their crosstalk in cancers.
Collapse
|
14
|
Papanicolaou M, Parker AL, Yam M, Filipe EC, Wu SZ, Chitty JL, Wyllie K, Tran E, Mok E, Nadalini A, Skhinas JN, Lucas MC, Herrmann D, Nobis M, Pereira BA, Law AMK, Castillo L, Murphy KJ, Zaratzian A, Hastings JF, Croucher DR, Lim E, Oliver BG, Mora FV, Parker BL, Gallego-Ortega D, Swarbrick A, O'Toole S, Timpson P, Cox TR. Temporal profiling of the breast tumour microenvironment reveals collagen XII as a driver of metastasis. Nat Commun 2022; 13:4587. [PMID: 35933466 PMCID: PMC9357007 DOI: 10.1038/s41467-022-32255-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/22/2022] [Indexed: 01/21/2023] Open
Abstract
The tumour stroma, and in particular the extracellular matrix (ECM), is a salient feature of solid tumours that plays a crucial role in shaping their progression. Many desmoplastic tumours including breast cancer involve the significant accumulation of type I collagen. However, recently it has become clear that the precise distribution and organisation of matrix molecules such as collagen I is equally as important in the tumour as their abundance. Cancer-associated fibroblasts (CAFs) coexist within breast cancer tissues and play both pro- and anti-tumourigenic roles through remodelling the ECM. Here, using temporal proteomic profiling of decellularized tumours, we interrogate the evolving matrisome during breast cancer progression. We identify 4 key matrisomal clusters, and pinpoint collagen type XII as a critical component that regulates collagen type I organisation. Through combining our proteomics with single-cell transcriptomics, and genetic manipulation models, we show how CAF-secreted collagen XII alters collagen I organisation to create a pro-invasive microenvironment supporting metastatic dissemination. Finally, we show in patient cohorts that collagen XII may represent an indicator of breast cancer patients at high risk of metastatic relapse. The distribution and organisation of matrix molecules in the tumour stroma help shape solid tumour progression. Here they perform temporal proteomic profiling of the matrisome during breast cancer progression and show that collagen XII secreted from CAFs provides a pro-invasive microenvironment.
Collapse
Affiliation(s)
- Michael Papanicolaou
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Amelia L Parker
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Michelle Yam
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Elysse C Filipe
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Sunny Z Wu
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Jessica L Chitty
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Kaitlin Wyllie
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Emmi Tran
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Ellie Mok
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Audrey Nadalini
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Joanna N Skhinas
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Morghan C Lucas
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - David Herrmann
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Max Nobis
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Brooke A Pereira
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew M K Law
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Lesley Castillo
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Kendelle J Murphy
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Anaiis Zaratzian
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Jordan F Hastings
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - David R Croucher
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Elgene Lim
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,Woolcock Institute of Medical Research, Respiratory Cellular and Molecular Biology, The University of Sydney, Sydney, NSW, Australia
| | - Fatima Valdes Mora
- Cancer Epigenetic Biology and Therapeutics, Personalised Medicine, Children's Cancer Institute, Sydney, NSW, 2031, Australia.,School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Benjamin L Parker
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - David Gallego-Ortega
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.,School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| | - Alexander Swarbrick
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Sandra O'Toole
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia.,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Paul Timpson
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia. .,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia. .,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| | - Thomas R Cox
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia. .,Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia. .,School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
15
|
Iori S, Pauletto M, Bassan I, Bonsembiante F, Gelain ME, Bardhi A, Barbarossa A, Zaghini A, Dacasto M, Giantin M. Deepening the Whole Transcriptomics of Bovine Liver Cells Exposed to AFB1: A Spotlight on Toll-like Receptor 2. Toxins (Basel) 2022; 14:toxins14070504. [PMID: 35878242 PMCID: PMC9323327 DOI: 10.3390/toxins14070504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 12/13/2022] Open
Abstract
Aflatoxin B1 (AFB1) is a food contaminant metabolized mostly in the liver and leading to hepatic damage. Livestock species are differently susceptible to AFB1, but the underlying mechanisms of toxicity have not yet been fully investigated, especially in ruminants. Thus, the aim of the present study was to better characterize the molecular mechanism by which AFB1 exerts hepatotoxicity in cattle. The bovine fetal hepatocyte cell line (BFH12) was exposed for 48 h to three different AFB1 concentrations (0.9 µM, 1.8 µM and 3.6 µM). Whole-transcriptomic changes were measured by RNA-seq analysis, showing significant differences in the expression of genes mainly involved in inflammatory response, oxidative stress, drug metabolism, apoptosis and cancer. As a confirmatory step, post-translational investigations on genes of interest were implemented. Cell death associated with necrosis rather than apoptosis events was noted. As far as the toxicity mechanism is concerned, a molecular pathway linking inflammatory response and oxidative stress was postulated. Toll-Like Receptor 2 (TLR2) activation, consequent to AFB1 exposure, triggers an intracellular signaling cascade involving a kinase (p38β MAPK), which in turn allows the nuclear translocation of the activator protein-1 (AP-1) and NF-κB, finally leading to the release of pro-inflammatory cytokines. Furthermore, a p38β MAPK negative role in cytoprotective genes regulation was postulated. Overall, our investigations improved the actual knowledge on the molecular effects of this worldwide relevant natural toxin in cattle.
Collapse
Affiliation(s)
- Silvia Iori
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, Legnaro, 35020 Padua, Italy; (S.I.); (M.P.); (I.B.); (F.B.); (M.E.G.); (M.D.)
| | - Marianna Pauletto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, Legnaro, 35020 Padua, Italy; (S.I.); (M.P.); (I.B.); (F.B.); (M.E.G.); (M.D.)
| | - Irene Bassan
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, Legnaro, 35020 Padua, Italy; (S.I.); (M.P.); (I.B.); (F.B.); (M.E.G.); (M.D.)
| | - Federico Bonsembiante
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, Legnaro, 35020 Padua, Italy; (S.I.); (M.P.); (I.B.); (F.B.); (M.E.G.); (M.D.)
- Department of Animal Medicine, Production and Health, University of Padua, Viale dell’Università 16, Legnaro, 35020 Padua, Italy
| | - Maria Elena Gelain
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, Legnaro, 35020 Padua, Italy; (S.I.); (M.P.); (I.B.); (F.B.); (M.E.G.); (M.D.)
| | - Anisa Bardhi
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Via Tolara di Sopra 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (A.B.); (A.B.); (A.Z.)
| | - Andrea Barbarossa
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Via Tolara di Sopra 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (A.B.); (A.B.); (A.Z.)
| | - Anna Zaghini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Via Tolara di Sopra 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (A.B.); (A.B.); (A.Z.)
| | - Mauro Dacasto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, Legnaro, 35020 Padua, Italy; (S.I.); (M.P.); (I.B.); (F.B.); (M.E.G.); (M.D.)
| | - Mery Giantin
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, Legnaro, 35020 Padua, Italy; (S.I.); (M.P.); (I.B.); (F.B.); (M.E.G.); (M.D.)
- Correspondence: ; Tel.: +39-049-827-2946
| |
Collapse
|
16
|
Tucker RP, Degen M. Revisiting the Tenascins: Exploitable as Cancer Targets? Front Oncol 2022; 12:908247. [PMID: 35785162 PMCID: PMC9248440 DOI: 10.3389/fonc.2022.908247] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
For their full manifestation, tumors require support from the surrounding tumor microenvironment (TME), which includes a specific extracellular matrix (ECM), vasculature, and a variety of non-malignant host cells. Together, these components form a tumor-permissive niche that significantly differs from physiological conditions. While the TME helps to promote tumor progression, its special composition also provides potential targets for anti-cancer therapy. Targeting tumor-specific ECM molecules and stromal cells or disrupting aberrant mesenchyme-cancer communications might normalize the TME and improve cancer treatment outcome. The tenascins are a family of large, multifunctional extracellular glycoproteins consisting of four members. Although each have been described to be expressed in the ECM surrounding cancer cells, tenascin-C and tenascin-W are currently the most promising candidates for exploitability and clinical use as they are highly expressed in various tumor stroma with relatively low abundance in healthy tissues. Here, we review what is known about expression of all four tenascin family members in tumors, followed by a more thorough discussion on tenascin-C and tenascin-W focusing on their oncogenic functions and their potential as diagnostic and/or targetable molecules for anti-cancer treatment purposes.
Collapse
Affiliation(s)
- Richard P. Tucker
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, CA, United States
| | - Martin Degen
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
- *Correspondence: Martin Degen,
| |
Collapse
|
17
|
Tognetti M, Sklodowski K, Müller S, Kamber D, Muntel J, Bruderer R, Reiter L. Biomarker Candidates for Tumors Identified from Deep-Profiled Plasma Stem Predominantly from the Low Abundant Area. J Proteome Res 2022; 21:1718-1735. [PMID: 35605973 PMCID: PMC9251764 DOI: 10.1021/acs.jproteome.2c00122] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
![]()
The plasma proteome
has the potential to enable a holistic analysis
of the health state of an individual. However, plasma biomarker discovery
is difficult due to its high dynamic range and variability. Here,
we present a novel automated analytical approach for deep plasma profiling
and applied it to a 180-sample cohort of human plasma from lung, breast,
colorectal, pancreatic, and prostate cancers. Using a controlled quantitative
experiment, we demonstrate a 257% increase in protein identification
and a 263% increase in significantly differentially abundant proteins
over neat plasma. In the cohort, we identified 2732 proteins. Using
machine learning, we discovered biomarker candidates such as STAT3
in colorectal cancer and developed models that classify the diseased
state. For pancreatic cancer, a separation by stage was achieved.
Importantly, biomarker candidates came predominantly from the low
abundance region, demonstrating the necessity to deeply profile because
they would have been missed by shallow profiling.
Collapse
Affiliation(s)
| | | | | | | | - Jan Muntel
- Biognosys, Schlieren, Zurich 8952, Switzerland
| | | | | |
Collapse
|
18
|
Morbid Obesity in Women Is Associated with an Altered Intestinal Expression of Genes Related to Cancer Risk and Immune, Defensive, and Antimicrobial Response. Biomedicines 2022; 10:biomedicines10051024. [PMID: 35625760 PMCID: PMC9138355 DOI: 10.3390/biomedicines10051024] [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: 04/06/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Little is known about the relation between morbid obesity and duodenal transcriptomic changes. We aimed to identify intestinal genes that may be associated with the development of obesity regardless of the degree of insulin resistance (IR) of patients. Material and Methods: Duodenal samples were assessed by microarray in three groups of women: non-obese women and women with morbid obesity with low and high IR. Results: We identified differentially expressed genes (DEGs) associated with morbid obesity, regardless of IR degree, related to digestion and lipid metabolism, defense response and inflammatory processes, maintenance of the gastrointestinal epithelium, wound healing and homeostasis, and the development of gastrointestinal cancer. However, other DEGs depended on the IR degree. We mainly found an upregulation of genes involved in the response to external organisms, hypoxia, and wound healing functions in women with morbid obesity and low IR. Conclusions: Regardless of the degree of IR, morbid obesity is associated with an altered expression of genes related to intestinal defenses, antimicrobial and immune responses, and gastrointestinal cancer. Our data also suggest a deficient duodenal immune and antimicrobial response in women with high IR.
Collapse
|
19
|
Ghoneum A, Almousa S, Warren B, Abdulfattah AY, Shu J, Abouelfadl H, Gonzalez D, Livingston C, Said N. Exploring the clinical value of tumor microenvironment in platinum-resistant ovarian cancer. Semin Cancer Biol 2021; 77:83-98. [PMID: 33476723 PMCID: PMC8286277 DOI: 10.1016/j.semcancer.2020.12.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/20/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022]
Abstract
Platinum resistance in epithelial ovarian cancer (OvCa) is rising at an alarming rate, with recurrence of chemo-resistant high grade serous OvCa (HGSC) in roughly 75 % of all patients. Additionally, HGSC has an abysmal five-year survival rate, standing at 39 % and 17 % for FIGO stages III and IV, respectively. Herein we review the crucial cellular interactions between HGSC cells and the cellular and non-cellular components of the unique peritoneal tumor microenvironment (TME). We highlight the role of the extracellular matrix (ECM), ascitic fluid as well as the mesothelial cells, tumor associated macrophages, neutrophils, adipocytes and fibroblasts in platinum-resistance. Moreover, we underscore the importance of other immune-cell players in conferring resistance, including natural killer cells, myeloid-derived suppressive cells (MDSCs) and T-regulatory cells. We show the clinical relevance of the key platinum-resistant markers and their correlation with the major pathways perturbed in OvCa. In parallel, we discuss the effect of immunotherapies in re-sensitizing platinum-resistant patients to platinum-based drugs. Through detailed analysis of platinum-resistance in HGSC, we hope to advance the development of more effective therapy options for this aggressive disease.
Collapse
Affiliation(s)
- Alia Ghoneum
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Sameh Almousa
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Bailey Warren
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Ammar Yasser Abdulfattah
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Alexandria University School of Medicine, Alexandria, Egypt
| | - Junjun Shu
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; The Third Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Hebatullah Abouelfadl
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Department of Genetics, Animal Health Research Institute, Dokki, Egypt
| | - Daniela Gonzalez
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Christopher Livingston
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Neveen Said
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Departments of Urology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Comprehensive Cancer Center, Winston Salem, NC, 27157, USA.
| |
Collapse
|
20
|
Fejza A, Poletto E, Carobolante G, Camicia L, Andreuzzi E, Capuano A, Pivetta E, Pellicani R, Colladel R, Marastoni S, Doliana R, Iozzo RV, Spessotto P, Mongiat M. Multimerin-2 orchestrates the cross-talk between endothelial cells and pericytes: A mechanism to maintain vascular stability. Matrix Biol Plus 2021; 11:100068. [PMID: 34435184 PMCID: PMC8377000 DOI: 10.1016/j.mbplus.2021.100068] [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] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
The ECM Multimerin-2 is a substrate for pericyte adhesion. The recruitment of pericytes leads to enhanced Multimerin-2 expression by endothelial cells. Multimerin-2 induces the expression of important cytokines both in endothelial cells and pericytes. The deposition of Multimerin-2 is key for the endothelial cell/pericyte crosstalk required for the establishment of vascular stability.
Tumor angiogenesis is vital for the growth and development of various solid cancers and as such is a valid and promising therapeutic target. Unfortunately, the use of the currently available anti-angiogenic drugs increases the progression-free survival by only a few months. Conversely, targeting angiogenesis to prompt both vessel reduction and normalization, has been recently viewed as a promising approach to improve therapeutic efficacy. As a double-edged sword, this line of attack may on one side halt tumor growth as a consequence of the reduction of nutrients and oxygen supplied to the tumor cells, and on the other side improve drug delivery and, hence, efficacy. Thus, it is of upmost importance to better characterize the mechanisms regulating vascular stability. In this context, recruitment of pericytes along the blood vessels is crucial to their maturation and stabilization. As the extracellular matrix molecule Multimerin-2 is secreted by endothelial cells and deposited also in juxtaposition between endothelial cells and pericytes, we explored Multimerin-2 role in the cross-talk between the two cell types. We discovered that Multimerin-2 is an adhesion substrate for pericytes. Interestingly, and consistent with the notion that Multimerin-2 is a homeostatic molecule deposited in the later stages of vessel formation, we found that the interaction between endothelial cells and pericytes promoted the expression of Multimerin-2. Furthermore, we found that Multimerin-2 modulated the expression of key cytokines both in endothelial cells and pericytes. Collectively, our findings posit Multimerin-2 as a key molecule in the cross-talk between endothelial cells and pericytes and suggest that the expression of this glycoprotein is required to maintain vascular stability.
Collapse
Key Words
- Ang-2, Angiopeietin-2
- Angiogenesis
- CD248, cluster of differentiation 248
- CD93, cluster of differentiation 93
- ECM, extracellular matrix
- EDEN, EMI Domain ENdowed
- Extracellular matrix
- HB-EGF, heparin binding epidermal growth factor
- HBVP, human brain vascular pericytes
- HDMEC, human dermal vascular endothelial cells
- HUVEC, human umbilical vein endothelial cells
- Notch-3-R, Notch Receptor 3
- PDGF, platelet-derived growth factor
- VEGFA, vascular endothelial growth factor A
- VEGFR2, vascular endothelial growth factor receptor 2
- VSMCs, vascular smooth muscle cells
- Vascular stability
Collapse
Affiliation(s)
- Albina Fejza
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Greta Carobolante
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Lucrezia Camicia
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Eva Andreuzzi
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Alessandra Capuano
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Eliana Pivetta
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Rosanna Pellicani
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Roberta Colladel
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Stefano Marastoni
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Roberto Doliana
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Paola Spessotto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| |
Collapse
|
21
|
Grill M, Lazzeri I, Kirsch A, Steurer N, Grossmann T, Karbiener M, Heitzer E, Gugatschka M. Vocal Fold Fibroblasts in Reinke's Edema Show Alterations Involved in Extracellular Matrix Production, Cytokine Response and Cell Cycle Control. Biomedicines 2021; 9:biomedicines9070735. [PMID: 34206882 PMCID: PMC8301432 DOI: 10.3390/biomedicines9070735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022] Open
Abstract
The voice disorder Reinke’s edema (RE) is a smoking- and voice-abuse associated benign lesion of the vocal folds, defined by an edema of the Reinke’s space, accompanied by pathological microvasculature changes and immune cell infiltration. Vocal fold fibroblasts (VFF) are the main cell type of the lamina propria and play a key role in the disease progression. Current therapy is restricted to symptomatic treatment. Hence, there is an urgent need for a better understanding of the molecular causes of the disease. In the present study, we investigated differential expression profiles of RE and control VFF by means of RNA sequencing. In addition, fast gene set enrichment analysis (FGSEA) was performed in order to obtain involved biological processes, mRNA and protein levels of targets of interest were further evaluated. We identified 74 differentially regulated genes in total, 19 of which were upregulated and 55 downregulated. Differential expression analysis and FGSEA revealed upregulated genes and pathways involved in extracellular matrix (ECM) remodeling, inflammation and fibrosis. Downregulated genes and pathways were involved in ECM degradation, cell cycle control and proliferation. The current study addressed for the first time a direct comparison of VFF from RE to control and evaluated immediate functional consequences.
Collapse
Affiliation(s)
- Magdalena Grill
- Division of Phoniatrics, Department of Otorhinolaryngology, Medical University of Graz, 8036 Graz, Austria; (M.G.); (N.S.); (T.G.); (M.K.); (M.G.)
| | - Isaac Lazzeri
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, 8010 Graz, Austria; (I.L.); (E.H.)
| | - Andrijana Kirsch
- Division of Phoniatrics, Department of Otorhinolaryngology, Medical University of Graz, 8036 Graz, Austria; (M.G.); (N.S.); (T.G.); (M.K.); (M.G.)
- Correspondence:
| | - Nina Steurer
- Division of Phoniatrics, Department of Otorhinolaryngology, Medical University of Graz, 8036 Graz, Austria; (M.G.); (N.S.); (T.G.); (M.K.); (M.G.)
| | - Tanja Grossmann
- Division of Phoniatrics, Department of Otorhinolaryngology, Medical University of Graz, 8036 Graz, Austria; (M.G.); (N.S.); (T.G.); (M.K.); (M.G.)
| | - Michael Karbiener
- Division of Phoniatrics, Department of Otorhinolaryngology, Medical University of Graz, 8036 Graz, Austria; (M.G.); (N.S.); (T.G.); (M.K.); (M.G.)
- Global Pathogen Safety, Baxter AG, (part of Takeda), 1220 Vienna, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, 8010 Graz, Austria; (I.L.); (E.H.)
| | - Markus Gugatschka
- Division of Phoniatrics, Department of Otorhinolaryngology, Medical University of Graz, 8036 Graz, Austria; (M.G.); (N.S.); (T.G.); (M.K.); (M.G.)
| |
Collapse
|
22
|
Aubert A, Mercier-Gouy P, Aguero S, Berthier L, Liot S, Prigent L, Alcaraz LB, Verrier B, Terreux R, Moali C, Lambert E, Valcourt U. Latent TGF-β Activation Is a Hallmark of the Tenascin Family. Front Immunol 2021; 12:613438. [PMID: 34054795 PMCID: PMC8155481 DOI: 10.3389/fimmu.2021.613438] [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] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor-β (TGF-β) isoforms are secreted as inactive complexes formed through non-covalent interactions between bioactive TGF-β entities and their N-terminal pro-domains called latency-associated peptides (LAP). Extracellular activation of latent TGF-β within this complex is a crucial step in the regulation of TGF-β activity for tissue homeostasis and immune cell function. We previously showed that the matrix glycoprotein Tenascin-X (TN-X) interacted with the small latent TGF-β complex and triggered the activation of the latent cytokine into a bioactive TGF-β. This activation most likely occurs through a conformational change within the latent TGF-β complex and requires the C-terminal fibrinogen-like (FBG) domain of the glycoprotein. As the FBG-like domain is highly conserved among the Tenascin family members, we hypothesized that Tenascin-C (TN-C), Tenascin-R (TN-R) and Tenascin-W (TN-W) might share with TN-X the ability to regulate TGF-β bioavailability through their C-terminal domain. Here, we demonstrate that purified recombinant full-length Tenascins associate with the small latent TGF-β complex through their FBG-like domains. This association promotes activation of the latent cytokine and subsequent TGF-β cell responses in mammary epithelial cells, such as cytostasis and epithelial-to-mesenchymal transition (EMT). Considering the pleiotropic role of TGF-β in numerous physiological and pathological contexts, our data indicate a novel common function for the Tenascin family in the regulation of tissue homeostasis under healthy and pathological conditions.
Collapse
Affiliation(s)
- Alexandre Aubert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Perrine Mercier-Gouy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Stéphanie Aguero
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Laurent Berthier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Sophie Liot
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Laura Prigent
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Lindsay B Alcaraz
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier (ICM), Montpellier, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Raphaël Terreux
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Catherine Moali
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Ulrich Valcourt
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| |
Collapse
|
23
|
Liot S, Balas J, Aubert A, Prigent L, Mercier-Gouy P, Verrier B, Bertolino P, Hennino A, Valcourt U, Lambert E. Stroma Involvement in Pancreatic Ductal Adenocarcinoma: An Overview Focusing on Extracellular Matrix Proteins. Front Immunol 2021; 12:612271. [PMID: 33889150 PMCID: PMC8056076 DOI: 10.3389/fimmu.2021.612271] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide and is predicted to become second in 2030 in industrialized countries if no therapeutic progress is made. Among the different types of pancreatic cancers, Pancreatic Ductal Adenocarcinoma (PDAC) is by far the most represented one with an occurrence of more than 90%. This specific cancer is a devastating malignancy with an extremely poor prognosis, as shown by the 5-years survival rate of 2–9%, ranking firmly last amongst all cancer sites in terms of prognostic outcomes for patients. Pancreatic tumors progress with few specific symptoms and are thus at an advanced stage at diagnosis in most patients. This malignancy is characterized by an extremely dense stroma deposition around lesions, accompanied by tissue hypovascularization and a profound immune suppression. Altogether, these combined features make access to cancer cells almost impossible for conventional chemotherapeutics and new immunotherapeutic agents, thus contributing to the fatal outcomes of the disease. Initially ignored, the Tumor MicroEnvironment (TME) is now the subject of intensive research related to PDAC treatment and could contain new therapeutic targets. In this review, we will summarize the current state of knowledge in the field by focusing on TME composition to understand how this specific compartment could influence tumor progression and resistance to therapies. Attention will be paid to Tenascin-C, a matrix glycoprotein commonly upregulated during cancer that participates to PDAC progression and thus contributes to poor prognosis.
Collapse
Affiliation(s)
- Sophie Liot
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Jonathan Balas
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Alexandre Aubert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Laura Prigent
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Perrine Mercier-Gouy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Philippe Bertolino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France
| | - Ana Hennino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France
| | - Ulrich Valcourt
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| |
Collapse
|
24
|
Tavernari D, Battistello E, Dheilly E, Petruzzella AS, Mina M, Sordet-Dessimoz J, Peters S, Krueger T, Gfeller D, Riggi N, Oricchio E, Letovanec I, Ciriello G. Nongenetic Evolution Drives Lung Adenocarcinoma Spatial Heterogeneity and Progression. Cancer Discov 2021; 11:1490-1507. [PMID: 33563664 DOI: 10.1158/2159-8290.cd-20-1274] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/21/2020] [Accepted: 01/22/2021] [Indexed: 11/16/2022]
Abstract
Cancer evolution determines molecular and morphologic intratumor heterogeneity and challenges the design of effective treatments. In lung adenocarcinoma, disease progression and prognosis are associated with the appearance of morphologically diverse tumor regions, termed histologic patterns. However, the link between molecular and histologic features remains elusive. Here, we generated multiomics and spatially resolved molecular profiles of histologic patterns from primary lung adenocarcinoma, which we integrated with molecular data from >2,000 patients. The transition from indolent to aggressive patterns was not driven by genetic alterations but by epigenetic and transcriptional reprogramming reshaping cancer cell identity. A signature quantifying this transition was an independent predictor of patient prognosis in multiple human cohorts. Within individual tumors, highly multiplexed protein spatial profiling revealed coexistence of immune desert, inflamed, and excluded regions, which matched histologic pattern composition. Our results provide a detailed molecular map of lung adenocarcinoma intratumor spatial heterogeneity, tracing nongenetic routes of cancer evolution. SIGNIFICANCE: Lung adenocarcinomas are classified based on histologic pattern prevalence. However, individual tumors exhibit multiple patterns with unknown molecular features. We characterized nongenetic mechanisms underlying intratumor patterns and molecular markers predicting patient prognosis. Intratumor patterns determined diverse immune microenvironments, warranting their study in the context of current immunotherapies.This article is highlighted in the In This Issue feature, p. 1307.
Collapse
Affiliation(s)
- Daniele Tavernari
- Swiss Cancer Center Leman, Lausanne, Switzerland.,Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Elena Battistello
- Swiss Cancer Center Leman, Lausanne, Switzerland.,Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research (ISREC), EPFL, Lausanne, Switzerland
| | - Elie Dheilly
- Swiss Cancer Center Leman, Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research (ISREC), EPFL, Lausanne, Switzerland
| | - Aaron S Petruzzella
- Swiss Cancer Center Leman, Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research (ISREC), EPFL, Lausanne, Switzerland
| | - Marco Mina
- Swiss Cancer Center Leman, Lausanne, Switzerland.,Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Solange Peters
- Swiss Cancer Center Leman, Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Thorsten Krueger
- Division of Thoracic Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - David Gfeller
- Swiss Cancer Center Leman, Lausanne, Switzerland.,Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Nicolo Riggi
- Swiss Cancer Center Leman, Lausanne, Switzerland.,Institute of Pathology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Elisa Oricchio
- Swiss Cancer Center Leman, Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research (ISREC), EPFL, Lausanne, Switzerland
| | - Igor Letovanec
- Swiss Cancer Center Leman, Lausanne, Switzerland. .,Institute of Pathology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Pathology, Central Institute, Hôpital du Valais, Sion, Switzerland
| | - Giovanni Ciriello
- Swiss Cancer Center Leman, Lausanne, Switzerland. .,Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
25
|
Wawrzyniak D, Grabowska M, Głodowicz P, Kuczyński K, Kuczyńska B, Fedoruk-Wyszomirska A, Rolle K. Down-regulation of tenascin-C inhibits breast cancer cells development by cell growth, migration, and adhesion impairment. PLoS One 2020; 15:e0237889. [PMID: 32817625 PMCID: PMC7440653 DOI: 10.1371/journal.pone.0237889] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
Tenascin-C (TNC) is an extracellular matrix (ECM) glycoprotein that plays an important role in cell proliferation, migration, and tumour invasion in various cancers. TNC is one of the main protein overexpressed in breast cancer, indicating a role for this ECM molecule in cancer pathology. In this study we have evaluated the TNC loss-off-function in breast cancer cells. In our approach, we used dsRNA sharing sequence homology with TNC mRNA, called ATN-RNA. We present the data showing the effects of ATN-RNA in MDA-MB-231 cells both in monolayer and three-dimensional culture. Cells treated with ATN-RNA were analyzed for phenotypic alterations in proliferation, migration, adhesion, cell cycle, multi-caspase activation and the involvement in epithelial to mesenchymal transition (EMT) processes. As complementary analysis the oncogenomic portals were used to assess the clinical implication of TNC expression on breast cancer patient's survival, showing the TNC overexpression associated with a poor survival outcome. Our approach applied first in brain tumors and then in breast cancer cell lines reveals that ATN-RNA significantly diminishes the cell proliferation, migration and additionally, reverses the mesenchymal cells phenotype to the epithelial one. Thus, TNC could be considered as the universal target in different types of tumors, where TNC overexpression is associated with poor prognosis.
Collapse
Affiliation(s)
- Dariusz Wawrzyniak
- Department of Molecular Neurooncology, Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Poznan, Poland
| | - Małgorzata Grabowska
- Department of Molecular Neurooncology, Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Poznan, Poland
| | - Paweł Głodowicz
- Department of Molecular Neurooncology, Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Poznan, Poland
| | - Konrad Kuczyński
- Department of Molecular Neurooncology, Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Poznan, Poland
- NanoBioMedical Centre, Adam Mickiewicz University, Poznan, Poland
| | - Bogna Kuczyńska
- Department of Molecular Neurooncology, Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Poznan, Poland
| | - Agnieszka Fedoruk-Wyszomirska
- Laboratory of Subcellular Structures Analysis, Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Poznan, Poland
| | - Katarzyna Rolle
- Department of Molecular Neurooncology, Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|