1
|
Viiklepp K, Knuutila JS, Nissinen L, Siljamäki E, Rappu P, Suwal U, Pellinen T, Kallajoki M, Meri S, Heino J, Kähäri VM, Riihilä P. Expression of C1q by macrophages and fibroblasts in tumor microenvironment is associated with progression and metastasis of cutaneous squamous cell carcinoma. J Invest Dermatol 2025:S0022-202X(25)00446-4. [PMID: 40311866 DOI: 10.1016/j.jid.2025.04.007] [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: 08/06/2024] [Revised: 03/10/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer, with poor prognosis for metastatic cases. We demonstrated previously that cSCC cells in culture express C1r and C1s components of the complement C1qr2s2 complex, but not C1q. Here, significantly higher mRNA levels of C1QA, C1QB, C1QC variants 1 and 2 were found in cSCC tumors compared to normal skin. Analysis of single cell RNA seq data of cSCC revealed expression of mRNAs for C1QA, C1QB, C1QC in macrophages and activated fibroblasts. C1q staining was detected on the surface of cSCC tumor cells, in peri- and intratumoral macrophages and in peritumoral activated fibroblasts using immunohistochemistry and multiplexed immunofluorescence. Expression was higher in cSCCs compared to normal skin, actinic keratoses, and cSCC in situ. C1q production was induced in 3D spheroid co-cultures of cSCC cells, fibroblasts, and macrophages. C1q stimulated growth of cSCC cells in culture. C1q expression was significantly more prevalent in metastatic primary cSCCs and in metastases compared to non-metastatic cSCCs. High C1q expression in cSCC correlated with poor prognosis. These findings provide evidence for macrophage- and fibroblast-derived C1q in the progression of cSCC. They also suggest stromal C1q as a marker for cSCC metastasis and poor prognosis.
Collapse
Affiliation(s)
- Kristina Viiklepp
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital,Turku, Finland
| | - Jaakko S Knuutila
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital,Turku, Finland
| | - Liisa Nissinen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital,Turku, Finland
| | - Elina Siljamäki
- MediCity Research Laboratory, University of Turku, FI-20520, Turku, Finland; Department of Life Technologies and InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Pekka Rappu
- MediCity Research Laboratory, University of Turku, FI-20520, Turku, Finland; Department of Life Technologies and InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Ujjwal Suwal
- MediCity Research Laboratory, University of Turku, FI-20520, Turku, Finland; Department of Life Technologies and InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland, FIMM, Helsinki Institute of Life Science (HiLIFE) University of Helsinki, Helsinki, Finland
| | - Markku Kallajoki
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Seppo Meri
- Department of Bacteriology and Immunology and the Translational Immunology Research Program, University of Helsinki, Finland
| | - Jyrki Heino
- MediCity Research Laboratory, University of Turku, FI-20520, Turku, Finland; Department of Life Technologies and InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital,Turku, Finland.
| | - Pilvi Riihilä
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital,Turku, Finland.
| |
Collapse
|
2
|
Heiskanen L, Nissinen L, Siljamäki E, Knuutila JS, Pellinen T, Kallajoki M, Heino J, Riihilä P, Kähäri VM. C5aR1 Promotes Invasion, Metastasis, and Poor Prognosis in Cutaneous Squamous Cell Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00073-2. [PMID: 40056975 DOI: 10.1016/j.ajpath.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 03/18/2025]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer, and the metastatic from is associated with a poor prognosis. Here, the role of the complement C5a receptor C5aR1 was examined in the progression and metastasis of cSCC. C5aR1 expression was increased in cSCC cells in a three-dimensional spheroid coculture model in the presence of fibroblasts, and treatment with recombinant C5a enhanced the invasion of cSCC cells. Staining for C5aR1 was detected on the surface of tumor cells at the invasive edge of human cSCC xenografts in vivo. Staining of metastatic and non-metastatic primary human cSCCs, premalignant and benign epidermal lesions, and normal skin for C5aR1 with multiplex immunofluorescence and chromogenic immunohistochemistry revealed increased expression of C5aR1 on the surface of tumor cells and fibroblasts in invasive cSCCs and recessive dystrophic epidermolysis bullosa-associated cSCCs compared with cSCC in situ, actinic keratoses, seborrheic keratoses, and normal skin. Increased expression of C5aR1 on the tumor cell surface and in fibroblasts was associated with metastatic risk and poor disease-specific survival of patients with primary cSCC. These findings suggest a role of C5a in cSCC cell invasion, and they identify C5aR1 as a novel biomarker for metastasis risk and poor prognosis in patients with cSCC. The results also suggest that C5aR1 could be a novel therapeutic target for the treatment of locally advanced and metastatic cSCC.
Collapse
Affiliation(s)
- Lauri Heiskanen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Liisa Nissinen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Elina Siljamäki
- Department of Life Technologies and InFLAMES Research Flagship, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Jaakko S Knuutila
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Markku Kallajoki
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Jyrki Heino
- Department of Life Technologies and InFLAMES Research Flagship, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Pilvi Riihilä
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland.
| |
Collapse
|
3
|
Li Z, Lu F, Zhou F, Song D, Chang L, Liu W, Yan G, Zhang G. From actinic keratosis to cutaneous squamous cell carcinoma: the key pathogenesis and treatments. Front Immunol 2025; 16:1518633. [PMID: 39925808 PMCID: PMC11802505 DOI: 10.3389/fimmu.2025.1518633] [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: 10/28/2024] [Accepted: 01/09/2025] [Indexed: 02/11/2025] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common non-melanoma skin cancer, among which 82% arise from actinic keratosis (AK) characterized by lesions of epidermal keratinocyte dysplasia. It is of great significance to uncover the progression mechanisms from AK to cSCC, which will facilitate the early therapeutic intervention of AK before malignant transformation. Thus, more and more studies are trying to ascertain the potential transformation mechanisms through multi-omics, including genetics, transcriptomics, and epigenetics. In this review, we gave an overview of the specific biomarkers and signaling pathways that may be involved in the pathogenesis from AK to cSCC, pointing out future possible molecular therapies for the early intervention of AK and cSCC. We also discussed current interventions on AK and cSCC, together with future perspectives.
Collapse
Affiliation(s)
- Zhenlin Li
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Fangqi Lu
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Fujin Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dekun Song
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lunhui Chang
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Weiying Liu
- Department of Dermatology, Hunan Aerospace Hospital, Changsha, China
| | - Guorong Yan
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Guolong Zhang
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Zabihi MR, Farhadi B, Akhoondian M. Complement protein expression changes in various conditions of breast cancer: in-silico analyses-experimental research. Ann Med Surg (Lond) 2024; 86:5152-5161. [PMID: 39239051 PMCID: PMC11374204 DOI: 10.1097/ms9.0000000000002216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 09/07/2024] Open
Abstract
Introduction Breast cancer is the most prevalent cancer diagnosed in females worldwide. The known biomarkers are insufficient to understand the actual prognosis of breast cancer, and identifying new biomarkers is desirable and valuable data to improve the patient's survival. Many inflammatory biomarkers, such as the complement system, can be regarded as prognostic values and as potent inflammatory mediators; complement proteins have a critical role in tumorigenesis. In the current study, the authors aim to investigate complement protein expression changes, particularly complement 3 (C3), complement 7 (C7), complement factor B (CFB), and complement factor D (CFD), in various conditions of breast cancer using in-silico tools. Methods The intent data were extracted using webtools, including; Kaplan-Meier plotter, BcGenExMiner, UALCAN, cbioportal, GeneMania, and Enrichr. To select valid data, a P greater than 0.05 was considered. Results The current study clarified that 21 complement genes correlated to survival conditions. Also, down or upregulation of extracted genes and breast cancer statuses were identified. Additionally, expression level difference of complement genes in various breast cancer four stages was detected. Ultimately, co-expression genes with complement genes were extracted and networked. Conclusion Changes in the expression of complement proteins can strongly correlate to breast cancer's prognosis, status, and survival. Furthermore, considering the vital role of CFD and CFB complement proteins in the alternative pathway in different stages of breast cancer, CFD and CFB can be regarded as reliable prognostic values for diagnosis.
Collapse
Affiliation(s)
- Mohammad Reza Zabihi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences
| | - Bahar Farhadi
- School of Medicine, Islamic Azad University, Mashhad Branch, Mashhad
| | - Mohammad Akhoondian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
5
|
Zhang T, Li Z, He A, Zhou W, Zhu X, Song Y. Clinical Significance and Potential Function of Complement Factor D in Acute Myeloid Leukemia. Cureus 2024; 16:e67260. [PMID: 39310420 PMCID: PMC11414840 DOI: 10.7759/cureus.67260] [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] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematologic malignancy characterized by aggressive proliferation and a poor prognosis. The objective of this study is to elucidate the specific role of complement factor D (CFD) in AML, with the aim of identifying robust prognostic markers for the disease. METHODS We performed a systematic investigation on clinical significance and potential function of CFD in AML by using the R Programming Language with The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), The Human Protein Atlas (HPA), The University of ALabama at Birmingham CANcer data analysis Portal (UALCAN), Gene Expression Profiling Interactive Analysis (GEPIA), Kaplan-Meier plotter, Cancer Cell Line Encyclopedia (CCLE) database, and Comprehensive Analysis on Multi-Omics of Immunotherapy in Pan-cancer (CAMOIP) database. The expression of CFD in AML patients was verified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). RESULTS The expression of CFD was the highest in AML cells than in other tumor cell lines. The expression of CFD was also higher in AML patients than in the matched normal group. Compared with the low expression of the CFD group, high expression of CFD predicted better overall survival (OS) and lower tumor mutational burden (TMB) in AML patients. Moreover, a nomogram model based on CFD was successfully constructed to predict the OS of AML patients. Notably, the expression of CFD was associated with drug sensitivity and monocyte cell infiltration. CONCLUSION CFD could serve as a potential OS prognostic biomarker and guide clinical treatment for AML.
Collapse
Affiliation(s)
- Taigang Zhang
- Department of Clinical Laboratory, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, CHN
| | - Zhaozhong Li
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, CHN
| | - Aoyu He
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, CHN
| | - Wenjuan Zhou
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, CHN
| | - Xianjin Zhu
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, CHN
| | - Yanfang Song
- Department of Clinical Laboratory, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, CHN
| |
Collapse
|
6
|
Nissinen L, Riihilä P, Viiklepp K, Rajagopal V, Storek MJ, Kähäri VM. C1s targeting antibodies inhibit the growth of cutaneous squamous carcinoma cells. Sci Rep 2024; 14:13465. [PMID: 38866870 PMCID: PMC11169539 DOI: 10.1038/s41598-024-64088-3] [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: 02/13/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer. The incidence of cSCC is increasing globally and the prognosis of metastatic disease is poor. Currently there are no specific targeted therapies for advanced or metastatic cSCC. We have previously shown abundant expression of the complement classical pathway C1 complex components, serine proteases C1r and C1s in tumor cells in invasive cSCCs in vivo, whereas the expression of C1r and C1s was lower in cSCCs in situ, actinic keratoses and in normal skin. We have also shown that knockdown of C1s expression results in decreased viability and growth of cSCC cells by promoting apoptosis both in culture and in vivo. Here, we have studied the effect of specific IgG2a mouse monoclonal antibodies TNT003 and TNT005 targeting human C1s in five primary non-metastatic and three metastatic cSCC cell lines that show intracellular expression of C1s and secretion of C1s into the cell culture media. Treatment of cSCC cells with TNT003 and TNT005 significantly inhibited their growth and viability and promoted apoptosis of cSCC cells. These data indicate that TNT003 and TNT005 inhibit cSCC cell growth in culture and warrant further investigation of C1s targeted inhibition in additional in vitro and in vivo models of cSCC.
Collapse
Affiliation(s)
- Liisa Nissinen
- Department of Dermatology and FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Hämeentie 11 TE6, 20520, Turku, Finland
| | - Pilvi Riihilä
- Department of Dermatology and FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Hämeentie 11 TE6, 20520, Turku, Finland
| | - Kristina Viiklepp
- Department of Dermatology and FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Hämeentie 11 TE6, 20520, Turku, Finland
| | | | | | - Veli-Matti Kähäri
- Department of Dermatology and FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Hämeentie 11 TE6, 20520, Turku, Finland.
| |
Collapse
|
7
|
Zhou S, Tao B, Guo Y, Gu J, Li H, Zou C, Tang S, Jiang S, Fu D, Li J. Integrating plasma protein-centric multi-omics to identify potential therapeutic targets for pancreatic cancer. J Transl Med 2024; 22:557. [PMID: 38858729 PMCID: PMC11165868 DOI: 10.1186/s12967-024-05363-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Deciphering the role of plasma proteins in pancreatic cancer (PC) susceptibility can aid in identifying novel targets for diagnosis and treatment. METHODS We examined the relationship between genetically determined levels of plasma proteins and PC through a systemic proteome-wide Mendelian randomization (MR) analysis utilizing cis-pQTLs from multiple centers. Rigorous sensitivity analyses, colocalization, reverse MR, replications with varying instrumental variable selections and additional datasets, as well as subsequent meta-analysis, were utilized to confirm the robustness of significant findings. The causative effect of corresponding protein-coding genes' expression and their expression pattern in single-cell types were then investigated. Enrichment analysis, between-protein interaction and causation, knock-out mice models, and mediation analysis with established PC risk factors were applied to indicate the pathogenetic pathways. These candidate targets were ultimately prioritized upon druggability and potential side effects predicted by a phenome-wide MR. RESULTS Twenty-one PC-related circulating proteins were identified in the exploratory phase with no evidence for horizontal pleiotropy or reverse causation. Of these, 11 were confirmed in a meta-analysis integrating external validations. The causality at a transcription level was repeated for neutrophil elastase, hydroxyacylglutathione hydrolase, lipase member N, protein disulfide-isomerase A5, xyloside xylosyltransferase 1. The carbohydrate sulfotransferase 11 and histo-blood group ABO system transferase exhibited high-support genetic colocalization evidence and were found to affect PC carcinogenesis partially through modulating body mass index and type 2 diabetes, respectively. Approved drugs have been established for eight candidate targets, which could potentially be repurposed for PC therapies. The phenome-wide investigation revealed 12 proteins associated with 51 non-PC traits, and interference on protein disulfide-isomerase A5 and cystatin-D would increase the risk of other malignancies. CONCLUSIONS By employing comprehensive methodologies, this study demonstrated a genetic predisposition linking 21 circulating proteins to PC risk. Our findings shed new light on the PC etiology and highlighted potential targets as priorities for future efforts in early diagnosis and therapeutic strategies of PC.
Collapse
Affiliation(s)
- Siyu Zhou
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Baian Tao
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yujie Guo
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jichun Gu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Hengchao Li
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Caifeng Zou
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Sichong Tang
- School of Medicine, Fudan University, Shanghai, 200240, China
| | - Shuheng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Deliang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Ji Li
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
8
|
Meri S, Magrini E, Mantovani A, Garlanda C. The Yin Yang of Complement and Cancer. Cancer Immunol Res 2023; 11:1578-1588. [PMID: 37902610 DOI: 10.1158/2326-6066.cir-23-0399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 10/31/2023]
Abstract
Cancer-related inflammation is a crucial component of the tumor microenvironment (TME). Complement activation occurs in cancer and supports the development of an inflammatory microenvironment. Complement has traditionally been considered a mechanism of immune resistance against cancer, and its activation is known to contribute to the cytolytic effects of antibody-based immunotherapeutic treatments. However, several studies have recently revealed that complement activation may exert protumoral functions by sustaining cancer-related inflammation and immunosuppression through different molecular mechanisms, targeting both the TME and cancer cells. These new discoveries have revealed that complement manipulation can be considered a new strategy for cancer therapies. Here we summarize our current understanding of the mechanisms by which the different elements of the complement system exert antitumor or protumor functions, both in preclinical studies and in human tumorigenesis. Complement components can serve as disease biomarkers for cancer stratification and prognosis and be exploited for tumor treatment.
Collapse
Affiliation(s)
- Seppo Meri
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University and University Hospital of Helsinki, Helsinki, Finland
| | | | - Alberto Mantovani
- IRCCS-Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Cecilia Garlanda
- IRCCS-Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
9
|
Kotol D, Woessmann J, Hober A, Álvez MB, Tran Minh KH, Pontén F, Fagerberg L, Uhlén M, Edfors F. Absolute Quantification of Pan-Cancer Plasma Proteomes Reveals Unique Signature in Multiple Myeloma. Cancers (Basel) 2023; 15:4764. [PMID: 37835457 PMCID: PMC10571728 DOI: 10.3390/cancers15194764] [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: 06/17/2023] [Revised: 08/31/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Mass spectrometry based on data-independent acquisition (DIA) has developed into a powerful quantitative tool with a variety of implications, including precision medicine. Combined with stable isotope recombinant protein standards, this strategy provides confident protein identification and precise quantification on an absolute scale. Here, we describe a comprehensive targeted proteomics approach to profile a pan-cancer cohort consisting of 1800 blood plasma samples representing 15 different cancer types. We successfully performed an absolute quantification of 253 proteins in multiplex. The assay had low intra-assay variability with a coefficient of variation below 20% (CV = 17.2%) for a total of 1013 peptides quantified across almost two thousand injections. This study identified a potential biomarker panel of seven protein targets for the diagnosis of multiple myeloma patients using differential expression analysis and machine learning. The combination of markers, including the complement C1 complex, JCHAIN, and CD5L, resulted in a prediction model with an AUC of 0.96 for the identification of multiple myeloma patients across various cancer patients. All these proteins are known to interact with immunoglobulins.
Collapse
Affiliation(s)
- David Kotol
- Science For Life Laboratory, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; (D.K.); (J.W.); (A.H.); (M.B.Á.); (K.H.T.M.); (L.F.); (M.U.)
- Department of Protein Science, Division of Systems Biology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Jakob Woessmann
- Science For Life Laboratory, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; (D.K.); (J.W.); (A.H.); (M.B.Á.); (K.H.T.M.); (L.F.); (M.U.)
- Department of Protein Science, Division of Systems Biology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Andreas Hober
- Science For Life Laboratory, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; (D.K.); (J.W.); (A.H.); (M.B.Á.); (K.H.T.M.); (L.F.); (M.U.)
- Department of Protein Science, Division of Systems Biology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - María Bueno Álvez
- Science For Life Laboratory, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; (D.K.); (J.W.); (A.H.); (M.B.Á.); (K.H.T.M.); (L.F.); (M.U.)
- Department of Protein Science, Division of Systems Biology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Khue Hua Tran Minh
- Science For Life Laboratory, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; (D.K.); (J.W.); (A.H.); (M.B.Á.); (K.H.T.M.); (L.F.); (M.U.)
- Department of Protein Science, Division of Systems Biology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Fredrik Pontén
- Rudbeck Laboratory, Uppsala University, 752 36 Uppsala, Sweden;
| | - Linn Fagerberg
- Science For Life Laboratory, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; (D.K.); (J.W.); (A.H.); (M.B.Á.); (K.H.T.M.); (L.F.); (M.U.)
- Department of Protein Science, Division of Systems Biology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Mathias Uhlén
- Science For Life Laboratory, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; (D.K.); (J.W.); (A.H.); (M.B.Á.); (K.H.T.M.); (L.F.); (M.U.)
- Department of Protein Science, Division of Systems Biology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Fredrik Edfors
- Science For Life Laboratory, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; (D.K.); (J.W.); (A.H.); (M.B.Á.); (K.H.T.M.); (L.F.); (M.U.)
- Department of Protein Science, Division of Systems Biology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
| |
Collapse
|
10
|
Concentration of Selected Adipokines and Factors Regulating Carbohydrate Metabolism in Patients with Head and Neck Cancer in Respect to Their Body Mass Index. Int J Mol Sci 2023; 24:ijms24043283. [PMID: 36834693 PMCID: PMC9959515 DOI: 10.3390/ijms24043283] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/29/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Head and neck cancers (HNCs) are a group of tumors not common in European populations. So far, not much is known about the role of obesity, adipokines, glucose metabolism, and inflammation in the pathogenesis of HNC. The aim of the study was to determine the concentrations of ghrelin, omentin-1, adipsin, adiponectin, leptin, resistin, visfatin, glucagon, insulin, C-peptide, glucagon-like peptide-1 (GLP-1), plasminogen activator inhibitor-1 (PAI-1), and gastric inhibitory peptide (GIP) in the blood serum of HNC patients depending on their body mass index (BMI). The study included 46 patients divided into two groups according to their BMI values: the normal BMI group (nBMI) included 23 patients with BMI < 25 kg/m2 and the increased BMI group (iBMI) included patients with BMI ≥ 25 kg/m2. A control group (CG) included 23 healthy people (BMI < 25 kg/m2). Statistically significant differences in the levels of adipsin, ghrelin, glucagon, PAI-1, and visfatin were shown between nBMI and CG. In the case of nBMI and iBMI, statistically significant differences were observed in the concentrations of adiponectin, C-peptide, ghrelin, GLP-1, insulin, leptin, omentin-1, PAI-1, resistin, and visfatin. The obtained results indicate a disruption of endocrine function of adipose tissue and impaired glucose metabolism in HNC. Obesity, which is not a typical risk factor for HNC, may aggravate the negative metabolic changes associated with this type of neoplasm. Ghrelin, visfatin, PAI-1, adipsin, and glucagon might be related to head and neck carcinogenesis. They seem to be promising directions for further research.
Collapse
|
11
|
Super Enhancer-Regulated LINC00094 (SERLOC) Upregulates the Expression of MMP-1 and MMP-13 and Promotes Invasion of Cutaneous Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14163980. [PMID: 36010973 PMCID: PMC9406669 DOI: 10.3390/cancers14163980] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as important regulators of cancer progression. Super enhancers (SE) play a role in tumorigenesis and regulate the expression of specific lncRNAs. We examined the role of BRD3OS, also named LINC00094, in cutaneous squamous cell carcinoma (cSCC). Elevated BRD3OS (LINC00094) expression was detected in cSCC cells, and expression was downregulated by SE inhibitors THZ1 and JQ1 and via the MEK1/ERK1/2 pathway. Increased expression of BRD3OS (LINC00094) was noted in tumor cells in cSCCs and their metastases compared to normal skin, actinic keratoses, and cSCCs in situ. Higher BRD3OS (LINC00094) expression was noted in metastatic cSCCs than in non-metastatic cSCCs. RNA-seq analysis after BRD3OS (LINC00094) knockdown revealed significantly regulated GO terms Cell-matrix adhesion, Basement membrane, Metalloendopeptidase activity, and KEGG pathway Extracellular matrix–receptor interaction. Among the top-regulated genes were MMP1, MMP10, and MMP13. Knockdown of BRD3OS (LINC00094) resulted in decreased production of MMP-1 and MMP-13 by cSCC cells, suppressed invasion of cSCC cells through collagen I, and growth of human cSCC xenografts in vivo. Based on these observations, BRD3OS (LINC00094) was named SERLOC (super enhancer and ERK1/2-Regulated Long Intergenic non-protein coding transcript Overexpressed in Carcinomas). These results reveal the role of SERLOC in cSCC invasion and identify it as a potential therapeutic target in advanced cSCC.
Collapse
|
12
|
Thind AS, Ashford B, Strbenac D, Mitchell J, Lee J, Mueller SA, Minaei E, Perry JR, Ch’ng S, Iyer NG, Clark JR, Gupta R, Ranson M. Whole genome analysis reveals the genomic complexity in metastatic cutaneous squamous cell carcinoma. Front Oncol 2022; 12:919118. [PMID: 35982973 PMCID: PMC9379253 DOI: 10.3389/fonc.2022.919118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/27/2022] [Indexed: 12/13/2022] Open
Abstract
Metastatic cutaneous squamous cell carcinoma (CSCC) is a highly morbid disease requiring radical surgery and adjuvant therapy, which is associated with a poor prognosis. Yet, compared to other advanced malignancies, relatively little is known of the genomic landscape of metastatic CSCC. We have previously reported the mutational signatures and mutational patterns of CCCTC-binding factor (CTCF) regions in metastatic CSCC. However, many other genomic components (indel signatures, non-coding drivers, and structural variants) of metastatic CSCC have not been reported. To this end, we performed whole genome sequencing on lymph node metastases and blood DNA from 25 CSCC patients with regional metastases of the head and neck. We designed a multifaceted computational analysis at the whole genome level to provide a more comprehensive perspective of the genomic landscape of metastatic CSCC. In the non-coding genome, 3′ untranslated region (3′UTR) regions of EVC (48% of specimens), PPP1R1A (48% of specimens), and ABCA4 (20% of specimens) along with the tumor-suppressing long non-coding RNA (lncRNA) LINC01003 (64% of specimens) were significantly functionally altered (Q-value < 0.05) and represent potential non-coding biomarkers of CSCC. Recurrent copy number loss in the tumor suppressor gene PTPRD was observed. Gene amplification was much less frequent, and few genes were recurrently amplified. Single nucleotide variants driver analyses from three tools confirmed TP53 and CDKN2A as recurrently mutated genes but also identified C9 as a potential novel driver in this disease. Furthermore, indel signature analysis highlighted the dominance of ID signature 13 (ID13) followed by ID8 and ID9. ID9 has previously been shown to have no association with skin melanoma, unlike ID13 and ID8, suggesting a novel pattern of indel variation in metastatic CSCC. The enrichment analysis of various genetically altered candidates shows enrichment of “TGF-beta regulation of extracellular matrix” and “cell cycle G1 to S check points.” These enriched terms are associated with genetic instability, cell proliferation, and migration as mechanisms of genomic drivers of metastatic CSCC.
Collapse
Affiliation(s)
- Amarinder Singh Thind
- School of Medicine, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| | - Bruce Ashford
- School of Medicine, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Illawarra Shoalhaven Local Health District, Wollongong, NSW, Australia
- *Correspondence: Bruce Ashford,
| | - Dario Strbenac
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, NSW, Australia
| | - Jenny Mitchell
- Illawarra Shoalhaven Local Health District, Wollongong, NSW, Australia
| | - Jenny Lee
- Sydney Head and Neck Cancer Institute, Chris O’Brien Lifehouse, Sydney, NSW, Australia
- Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Simon A. Mueller
- Sydney Head and Neck Cancer Institute, Chris O’Brien Lifehouse, Sydney, NSW, Australia
- Department of Otorhinolaryngology, Head and Neck Surgery, Zurich University Hospital and University of Zurich, Zurich, Switzerland
| | - Elahe Minaei
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Jay R. Perry
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Sydney Ch’ng
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, NSW, Australia
- Sydney Head and Neck Cancer Institute, Chris O’Brien Lifehouse, Sydney, NSW, Australia
| | - N. Gopalakrishna Iyer
- Department of Head and Neck Surgery, National Cancer Center, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Jonathan R. Clark
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, NSW, Australia
- Sydney Head and Neck Cancer Institute, Chris O’Brien Lifehouse, Sydney, NSW, Australia
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Sydney, NSW, Australia
| | - Ruta Gupta
- Anatomical Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
13
|
Quadri M, Marconi A, Sandhu SK, Kiss A, Efimova T, Palazzo E. Investigating Cutaneous Squamous Cell Carcinoma in vitro and in vivo: Novel 3D Tools and Animal Models. Front Med (Lausanne) 2022; 9:875517. [PMID: 35646967 PMCID: PMC9131878 DOI: 10.3389/fmed.2022.875517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/19/2022] [Indexed: 12/07/2022] Open
Abstract
Cutaneous Squamous Cell Carcinoma (cSCC) represents the second most common type of skin cancer, which incidence is continuously increasing worldwide. Given its high frequency, cSCC represents a major public health problem. Therefore, to provide the best patients’ care, it is necessary having a detailed understanding of the molecular processes underlying cSCC development, progression, and invasion. Extensive efforts have been made in developing new models allowing to study the molecular pathogenesis of solid tumors, including cSCC tumors. Traditionally, in vitro studies were performed with cells grown in a two-dimensional context, which, however, does not represent the complexity of tumor in vivo. In the recent years, new in vitro models have been developed aiming to mimic the three-dimensionality (3D) of the tumor, allowing the evaluation of tumor cell-cell and tumor-microenvironment interaction in an in vivo-like setting. These models include spheroids, organotypic cultures, skin reconstructs and organoids. Although 3D models demonstrate high potential to enhance the overall knowledge in cancer research, they lack systemic components which may be solved only by using animal models. Zebrafish is emerging as an alternative xenotransplant model in cancer research, offering a high-throughput approach for drug screening and real-time in vivo imaging to study cell invasion. Moreover, several categories of mouse models were developed for pre-clinical purpose, including xeno- and syngeneic transplantation models, autochthonous models of chemically or UV-induced skin squamous carcinogenesis, and genetically engineered mouse models (GEMMs) of cSCC. These models have been instrumental in examining the molecular mechanisms of cSCC and drug response in an in vivo setting. The present review proposes an overview of in vitro, particularly 3D, and in vivo models and their application in cutaneous SCC research.
Collapse
Affiliation(s)
- Marika Quadri
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Marconi
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Simran K Sandhu
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,The George Washington Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Alexi Kiss
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,The George Washington Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Tatiana Efimova
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,The George Washington Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Elisabetta Palazzo
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|