1
|
Li G, Mao Y, Liao J, Zhou Y. Integrated multiomics and Mendelian randomization identify CHIT1 as a novel sepsis biomarker and therapeutic target. Sci Rep 2025; 15:15715. [PMID: 40325173 PMCID: PMC12052846 DOI: 10.1038/s41598-025-99619-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/21/2025] [Indexed: 05/07/2025] Open
Abstract
Sepsis is characterized by severe organ failure due to an impaired response to infection. The underlying pathophysiology of sepsis is characterized by concurrent unbalanced hyperinflammatory and immunoparalysis. This study aimed to identify new key biomarkers that could predict outcomes in sepsis patients and explore theirunderlying molecular mechanisms. Bulk transcriptome data (GSE65682, GSE28750, GSE57065, GSE95233) and scRNA-seq data (GSE167363) of sepsis were obtained from the GEO database. Data for MR analysis were sourced from the eQTLGen Consortium and IEU OpenGWAS project. Prognostic biomarkers and potential drug targets for sepsis were identified through univariate Cox regression and MR analysis. The expression of these biomarkers was further validated using scRNA-seq data to investigate the underlying molecular mechanisms. Significantly higher expression of CHIT1 was found at sepsis non-survivor and associated with 28-day mortality of sepsis. scRNA-seq data of septic samples found that CHIT1 mainly expressed in neutrophils, which was also higher in sepsis non-survivors. The CHIT1 + neutrophils expressed higher inflammation related genes of S100A8, S100A9, S100A11, S100A12, IL1R2, IFNGR2, TLR2 and CXCL8 and reduced expression of HLA related genes of HLA-DMA, HLA-DPA1, HLA-DPB1, HLA-DRA, HLA-DRB1 and HLA-DRB5. Moreover, cell-chat analysis also showed that CHIT1 + neutrophils could interact with other immune cell types, including NK cells, erythroid cells, monocytes/macrophages, and DC by the way of ICAM1-(ITGAM + ITGB2) pathway. We identified CHIT1 as new biomarker and potential drug target for sepsis, which may intensify hyperinflammation and immune suppression of neutrophils. Developing immunotherapeutic strategies aimed at targeting CHIT1 would help to enhance sepsis outcomes.
Collapse
Affiliation(s)
- Guorui Li
- Department of Gerontology, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, 511300, Guangdong, China
| | - Yunlong Mao
- Department of Gerontology, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, 511300, Guangdong, China
| | - Jiaxiang Liao
- Department of Emergence, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuquan Zhou
- Department of Gerontology, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, 511300, Guangdong, China.
| |
Collapse
|
2
|
Majumder S, Mishra S, Shinde N, Cuitino MC, Bauer M, Ahirwar D, Basree MM, Bharti V, Ormiston K, Mawalkar R, Alsammerai S, Sarathy G, Vilgelm AE, Zhang X, Ganju RK, Ramaswamy B. Divergent paths of mammary gland involution: unveiling the cellular dynamics in abruptly and gradually involuted mouse models. Breast Cancer Res 2025; 27:1. [PMID: 39754221 PMCID: PMC11697808 DOI: 10.1186/s13058-024-01933-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Epidemiological studies associate an increase in breast cancer risk, particularly triple-negative breast cancer (TNBC), with lack of breastfeeding. This is more prevalent in African American women, with significantly lower rate of breastfeeding compared to Caucasian women. Prolonged breastfeeding leads to gradual involution (GI), whereas short-term or lack of breastfeeding leads to abrupt involution (AI) of the breast. Our previous study utilizing a murine model demonstrated precancerous changes, specifically hyperplasia, a non-obligate precursor of breast cancer in the mammary glands of AI mice. Here we investigated mechanisms during early events of AI that prompts precancerous changes in mouse mammary glands. METHODS Uniparous FVB/N mice were randomized to AI and GI on postpartum day 7 when all pups were removed from AI dams. GI dams were allowed to nurse the pups till day 31. Cell death kinetics and gene expression were assessed by TUNEL assay and qPCR respectively. Immune cell changes were investigated by flow cytometry, cytokine array and multiplex immunofluorescence. 3D-organoid cultures were used for in vitro assay of luminal progenitor cells. RESULTS AI results in rapid cell death, DNA repair response, and immunosuppressive myeloid cells infiltration, leading to a chronically inflamed microenvironment. GI elicits a more controlled immune response and extended cell death. At the peak of cell death, AI glands harbored more immunosuppressive myeloid-derived suppressor cells (MDSCs) and CD206 + M2-like macrophages, known to promote oncogenic events, compared to GI glands. AI glands exhibit an enrichment of CCL9-producing MDSCs and CD206 + M2-like macrophages that promote expansion of ELF5 + /ERα- luminal cells, both in vitro and in vivo. Multiplex imaging of AI glands demonstrated an increase in ELF5 + /WNT5a + luminal cells alongside a reduction in the ELF5 + /ERα + population when involution appeared histologically complete. A significantly higher number of CD206 + cells in post involution AI gland attests to a chronically inflamed state induced by AI. CONCLUSIONS Our findings reveal significant disparities between AI and GI gland dynamics at the early phase of involution. CCL9, secreted by immune cells at the peak of cell death promotes expansion of Elf5 + /ERα- luminal progenitor cells, the putative precursors of TNBC connecting early events of AI with increased breast cancer risk.
Collapse
Affiliation(s)
- Sarmila Majumder
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| | - Sanjay Mishra
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Neelam Shinde
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Maria C Cuitino
- Department of Radiation Oncology, The Ohio State University, Columbus, OH, USA
| | - Morgan Bauer
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Dinesh Ahirwar
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Mustafa M Basree
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Vijaya Bharti
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Kate Ormiston
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Resham Mawalkar
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Sara Alsammerai
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gautam Sarathy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Anna E Vilgelm
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Xiaoli Zhang
- Department of Biomedical Informatics and Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Ramesh K Ganju
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
3
|
Su PC, Chen CY, Yu MH, Kuo IY, Yang PS, Hsu CH, Hou YC, Hsieh HT, Chang CP, Shan YS, Wang YC. Fully human chitinase-3 like-1 monoclonal antibody inhibits tumor growth, fibrosis, angiogenesis, and immune cell remodeling in lung, pancreatic, and colorectal cancers. Biomed Pharmacother 2024; 176:116825. [PMID: 38820971 DOI: 10.1016/j.biopha.2024.116825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024] Open
Abstract
Considering the limited efficacy of current therapies in lung, colorectal, and pancreatic cancers, innovative combination treatments with diverse mechanisms of action are needed to improve patients' outcomes. Chitinase-3 like-1 protein (CHI3L1) emerges as a versatile factor with significant implications in various diseases, particularly cancers, fostering an immunosuppressive tumor microenvironment for cancer progression. Therefore, pre-clinical validation is imperative to fully realize its potential in cancer treatment. We developed phage display-derived fully human monoclonal CHI3L1 neutralizing antibodies (nAbs) and verified the nAbs-antigen binding affinity and specificity in lung, pancreatic and colorectal cancer cell lines. Tumor growth signals, proliferation and migration ability were all reduced by CHI3L1 nAbs in vitro. Orthotopic or subcutaneous tumor mice model and humanized mouse model were established for characterizing the anti-tumor properties of two CHI3L1 nAb leads. Importantly, CHI3L1 nAbs not only inhibited tumor growth but also mitigated fibrosis, angiogenesis, and restored immunostimulatory functions of immune cells in pancreatic, lung, and colorectal tumor mice models. Mechanistically, CHI3L1 nAbs directly suppressed the activation of pancreatic stellate cells and the transformation of macrophages into myofibroblasts, thereby attenuating fibrosis. These findings strongly support the therapeutic potential of CHI3L1 nAbs in overcoming clinical challenges, including the failure of gemcitabine in pancreatic cancer.
Collapse
Affiliation(s)
- Pei-Chia Su
- Department of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - Ching-Yu Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - Min-Hua Yu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - I-Ying Kuo
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, No.100, Shih-Chuan 1st Road, Kaohsiung 80708, Taiwan
| | - Pei-Shan Yang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - Ching-Hsuan Hsu
- AP Biosciences, Inc, No. 508, Sec. 7, Zhongxiao E. Rd, Taipei 115011, Taiwan
| | - Ya-Chin Hou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No.138, Sheng-Li Road, Tainan 70403, Taiwan; Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No.138, Sheng-Li Road, Tainan 70403, Taiwan
| | - Hsin-Ta Hsieh
- AP Biosciences, Inc, No. 508, Sec. 7, Zhongxiao E. Rd, Taipei 115011, Taiwan
| | - Chih-Peng Chang
- Department of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan; Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No.138, Sheng-Li Road, Tainan 70403, Taiwan; Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No.138, Sheng-Li Road, Tainan 70403, Taiwan.
| | - Yi-Ching Wang
- Department of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan.
| |
Collapse
|
4
|
Li D, Fan G, Zhou Y. Chitinase 3 like-1 activates the Akt pathway, inducing NF-κB-dependent release of pro-inflammatory cytokines and promoting the proliferative ability in nasopharyngeal carcinoma cells. Cytokine 2024; 179:156631. [PMID: 38710115 DOI: 10.1016/j.cyto.2024.156631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Chitinase 3 like-1 (CHI3L1) has been reported to function as an oncogene in many types of cancer. However, the biological function of CHI3L1 in nasopharyngeal carcinoma (NPC) remains unknown. METHODS Differentially expressed genes (DEGs) in NPC tissues in GSE64634 and GSE12452 were downloaded from Gene Expression Omnibus (GEO). CHI3L1, interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α) mRNA expression was examined by qRT-PCR. Cell proliferation was evaluated by CCK-8 and EdU incorporation assays. Western blot analysis was used to measure the changes of CHI3L1, nuclear factor-κappaB (NF-κB), and protein kinase B (Akt) pathways. Gene ontology (GO) enrichment and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway analyses were performed using DAVID database. RESULTS We identified 3 overlapping DEGs using Draw Venn diagram, among which CHI3L1 was chosen for the following analyses. CHI3L1 was upregulated in NPC tissues and cells. CHI3L1 silencing suppressed inflammatory response by inactivating the NF-κB pathway and inhibited cell proliferation in NPC cells. On the contrary, CHI3L1 overexpression induced inflammatory response by activating the NF-κB pathway and promoted cell proliferation in NPC cells. According to GO and KEGG analyses, CHI3L1 positive regulates Akt signaling and is enriched in the PI3K-Akt pathway. CHI3L1 knockdown inhibited the Akt pathway, and CHI3L1 overexpression activated the Akt pathway in NPC cells. Akt overexpression abolished the effects of CHI3L1 knockdown on inflammatory response, NF-κB pathway, and proliferation in NPC cells. On the contrary, Akt knockdown abolished the effects of CHI3L1 overexpression on inflammatory response, NF-κB pathway, and proliferation in NPC cells. CONCLUSION CHI3L1 knockdown inhibited NF-κB-dependent inflammatory response and promoting proliferation in NPC cells by inactivating the Akt pathway.
Collapse
Affiliation(s)
- Dajun Li
- Department of ENT, Nanyang First People's Hospital, Nanyang, China.
| | - Gai Fan
- Department of ENT, Nanyang First People's Hospital, Nanyang, China
| | - Yeqi Zhou
- Department of Radiotherapy, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
| |
Collapse
|
5
|
Wang Y, Zhong F, Xiao F, Li J, Liu X, Ni G, Wang T, Zhang W. Host-defence caerin 1.1 and 1.9 peptides suppress glioblastoma U87 and U118 cell proliferation through the modulation of mitochondrial respiration and induce the downregulation of CHI3L1. PLoS One 2024; 19:e0304149. [PMID: 38848430 PMCID: PMC11161062 DOI: 10.1371/journal.pone.0304149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Abstract
Glioblastoma, the most aggressive form of brain cancer, poses a significant global health challenge with a considerable mortality rate. With the predicted increase in glioblastoma incidence, there is an urgent need for more effective treatment strategies. In this study, we explore the potential of caerin 1.1 and 1.9, host defence peptides derived from an Australian tree frog, in inhibiting glioblastoma U87 and U118 cell growth. Our findings demonstrate the inhibitory impact of caerin 1.1 and 1.9 on cell growth through CCK8 assays. Additionally, these peptides effectively curtail the migration of glioblastoma cells in a cell scratch assay, exhibiting varying inhibitory effects among different cell lines. Notably, the peptides hinder the G0/S phase replication in both U87 and U118 cells, pointing to their impact on the cell cycle. Furthermore, caerin 1.1 and 1.9 show the ability to enter the cytoplasm of glioblastoma cells, influencing the morphology of mitochondria. Proteomics experiments reveal intriguing insights, with a decrease in CHI3L1 expression and an increase in PZP and JUNB expression after peptide treatment. These proteins play roles in cell energy metabolism and inflammatory response, suggesting a multifaceted impact on glioblastoma cells. In conclusion, our study underscores the substantial anticancer potential of caerin 1.1 and 1.9 against glioblastoma cells. These findings propose the peptides as promising candidates for further exploration in the realm of glioblastoma management, offering new avenues for developing effective treatment strategies.
Collapse
Affiliation(s)
- Yichen Wang
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
| | - Furong Zhong
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Zhongˈao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, China
| | - Fengyun Xiao
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
| | - Junjie Li
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Zhongˈao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, China
| | - Xiaosong Liu
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Zhongˈao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, China
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Guoying Ni
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Zhongˈao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, China
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Tianfang Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore BC, QLD, Australia
| | - Wei Zhang
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, Guangdong, China
- Guangdong Provincial Engineering and Technology Research Center of Stem Cell Therapy for Pituitary Disease, Guangzhou, China
| |
Collapse
|
6
|
Qiu K, Ding D, Zhang F, Yang B. LTF as a Potential Prognostic and Immunological Biomarker in Glioblastoma. Biochem Genet 2024:10.1007/s10528-024-10716-6. [PMID: 38763993 DOI: 10.1007/s10528-024-10716-6] [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: 10/24/2023] [Accepted: 01/24/2024] [Indexed: 05/21/2024]
Abstract
The lactoferrin (LTF) gene behaves like a tumor suppressor gene in diverse tumors, such as renal cancer, nasopharyngeal carcinoma and gastric cancer. However, the prognostic value of LTF expression in patients with glioblastoma remains unclear. In this study, the expression levels of LTF in patients with GBM were investigated in TCGA, GEPIA, CGGA and GEO database, and a survival analysis of LTF based on TCGA and CGGA was performed. Furthermore, the present study demonstrated the LTF gene co-expression, PPI network, KEGG/GO enrichment and immune cell infiltration analysis on TCGA and TIMER2.0 database. We found that LTF expression was significantly upregulated in GBM samples compared with normal samples and other glioma samples, and Kaplan-Meier analysis demonstrated that the overexpression of LTF were significantly associated with worse overall survival (OS) and 5-year OS in GBM patients (P < 0.05). KEGG/GO enrichment analysis demonstrated that functions of LTF concentrated in immune and inflammatory response and peptidase regulation (P < 0.05). Immune cell infiltration analysis presented that high LTF expression exhibited dysregulated immune infiltration (i.e., CD4 + T cells, neutrophils, macrophages, myeloid dendritic cells and cancer associated fibroblast). LTF was upregulated in tumors and correlated with worse OS in GBM patients, and LTF might function as an oncogene via inducing dysregulated immune infiltration in GBM.
Collapse
Affiliation(s)
- Kai Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Daling Ding
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Fengjiang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China.
| |
Collapse
|
7
|
Jung M, Bonavida B. Immune Evasion in Cancer Is Regulated by Tumor-Asociated Macrophages (TAMs): Targeting TAMs. Crit Rev Oncog 2024; 29:1-17. [PMID: 38989734 DOI: 10.1615/critrevoncog.2024053096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Recent advancements in cancer treatment have explored a variety of approaches to address the needs of patients. Recently, immunotherapy has evolved as an efficacious treatment for various cancers resistant to conventional therapies. Hence, significant milestones in immunotherapy were achieved clinically in a large subset of cancer patients. Unfortunately, some cancer types do not respond to treatment, and among the responsive cancers, some patients remain unresponsive to treatment. Consequently, there is a critical need to examine the mechanisms of immune resistance and devise strategies to target immune suppressor cells or factors, thereby allowing for tumor sensitivity to immune cytotoxic cells. M2 macrophages, also known as tumor-associated macrophages (TAMs), are of interest due to their role in suppressing the immune system and influencing antitumor immune responses through modulating T cell activity and immune checkpoint expression. TAMs are associated with signaling pathways that modulate the tumor microenvironment (TME), contributing to immune evasion. One approach targets TAMs, focusing on preventing the polarization of M1 macrophages into the protumoral M2 phenotype. Other strategies focus on direct or indirect targeting of M2 macrophages through understanding the interaction of TAMs with immune factors or signaling pathways. Clinically, biomarkers associated with TAMs' immune resistance in cancer patients have been identified, opening avenues for intervention using pharmacological agents or immunotherapeutic approaches. Ultimately, these multifaceted approaches are promising in overcoming immune resistance and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Megan Jung
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
8
|
Larionova I, Tashireva L. Immune gene signatures as prognostic criteria for cancer patients. Ther Adv Med Oncol 2023; 15:17588359231189436. [PMID: 37547445 PMCID: PMC10399276 DOI: 10.1177/17588359231189436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023] Open
Abstract
Recently, the possibility of using immune gene signatures (IGSs) has been considered as a novel prognostic tool for numerous cancer types. State-of-the-art methods of genomic, transcriptomic, and protein analysis have allowed the identification of a number of immune signatures correlated to disease outcome. The major adaptive and innate immune components are the T lymphocytes and macrophages, respectively. Herein, we collected essential data on IGSs consisting of subsets of T cells and tumor-associated macrophages and indicating cancer patient outcomes. We discuss factors that can introduce errors in the recognition of immune cell types and explain why the significance of immune signatures can be interpreted with uncertainty. The unidirectional functions of cell types should be entirely addressed in the signatures constructed by the combination of innate and adaptive immune cells. The state of the antitumor immune response is the key basis for IGSs and should be considered in gene signature construction. We also analyzed immune signatures for the prediction of immunotherapy response. Finally, we attempted to explain the present-day limitations in the use of immune signatures as robust criteria for prognosis.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 36 Lenina Av., Tomsk 634050, Russia
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Liubov Tashireva
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
9
|
Kim D, An L, Moon J, Maymi VI, McGurk AI, Rudd BD, Fowell DJ, White AC. Ccr2+ Monocyte-Derived Macrophages Influence Trajectories of Acquired Therapy Resistance in Braf-Mutant Melanoma. Cancer Res 2023; 83:2328-2344. [PMID: 37195124 PMCID: PMC10478295 DOI: 10.1158/0008-5472.can-22-2841] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/12/2023] [Accepted: 05/12/2023] [Indexed: 05/18/2023]
Abstract
Therapies targeting oncogene addiction have had a tremendous impact on tumor growth and patient outcome, but drug resistance continues to be problematic. One approach to deal with the challenge of resistance entails extending anticancer treatments beyond targeting cancer cells by additionally altering the tumor microenvironment. Understanding how the tumor microenvironment contributes to the evolution of diverse resistance pathways could aid in the design of sequential treatments that can elicit and take advantage of a predictable resistance trajectory. Tumor-associated macrophages often support neoplastic growth and are frequently the most abundant immune cell found in tumors. Here, we used clinically relevant in vivo Braf-mutant melanoma models with fluorescent markers to track the stage-specific changes in macrophages under targeted therapy with Braf/Mek inhibitors and assessed the dynamic evolution of the macrophage population generated by therapy pressure-induced stress. During the onset of a drug-tolerant persister state, Ccr2+ monocyte-derived macrophage infiltration rose, suggesting that macrophage influx at this point could facilitate the onset of stable drug resistance that melanoma cells show after several weeks of treatment. Comparison of melanomas that develop in a Ccr2-proficient or -deficient microenvironment demonstrated that lack of melanoma infiltrating Ccr2+ macrophages delayed onset of resistance and shifted melanoma cell evolution towards unstable resistance. Unstable resistance was characterized by sensitivity to targeted therapy when factors from the microenvironment were lost. Importantly, this phenotype was reversed by coculturing melanoma cells with Ccr2+ macrophages. Overall, this study demonstrates that the development of resistance may be directed by altering the tumor microenvironment to improve treatment timing and the probability of relapse. SIGNIFICANCE Ccr2+ melanoma macrophages that are active in tumors during the drug-tolerant persister state following targeted therapy-induced regression are key contributors directing melanoma cell reprogramming toward specific therapeutic resistance trajectories.
Collapse
Affiliation(s)
- Dahihm Kim
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853
| | - Luye An
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853
| | - Jiwon Moon
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853
| | - Viviana I Maymi
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Alexander I McGurk
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Deborah J Fowell
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Andrew C White
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853
| |
Collapse
|
10
|
Hong DE, Yu JE, Yoo SS, Yeo IJ, Son DJ, Yun J, Han SB, Hong JT. CHI3L1 induces autophagy through the JNK pathway in lung cancer cells. Sci Rep 2023; 13:9964. [PMID: 37340009 DOI: 10.1038/s41598-023-36844-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/11/2023] [Indexed: 06/22/2023] Open
Abstract
CHI3L1 is closely related to the molecular mechanisms of cancer cell migration, growth, and death. According to recent research, autophagy regulates tumor growth during various stages of cancer development. This study examined the association between CHI3L1 and autophagy in human lung cancer cells. In CHI3L1-overexpressing lung cancer cells, the expression of LC3, an autophagosome marker, and the accumulation of LC3 puncta increased. In contrast, CHI3L1 depletion in lung cancer cells decreased the formation of autophagosomes. Additionally, CHI3L1 overexpression promoted the formation of autophagosomes in various cancer cell lines: it also increased the co-localization of LC3 and the lysosome marker protein LAMP-1, indicating an increase in the production of autolysosomes. In mechanism study, CHI3L1 promotes autophagy via activation of JNK signaling. JNK may be crucial for CHI3L1-induced autophagy since pretreatment with the JNK inhibitor reduced the autophagic effect. Consistent with the in vitro model, the expression of autophagy-related proteins was downregulated in the tumor tissues of CHI3L1-knockout mice. Furthermore, the expression of autophagy-related proteins and CHI3L1 increased in lung cancer tissues compared with normal lung tissues. These findings show that CHI3L1-induced autophagy is triggered by JNK signals and that CHI3L1-induced autophagy could be a novel therapeutic approach to lung cancer.
Collapse
Affiliation(s)
- Da Eun Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Ji Eun Yu
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Seung Sik Yoo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
11
|
Weckman AM, Elphinstone RE, Ssenkusu JM, Tran V, Zhong K, Madanitsa M, Khairallah C, Kalilani-Phiri L, Mwapasa V, Conroy AL, Ter Kuile FO, McDonald CR, Kain KC. Sequential disruptions to inflammatory and angiogenic pathways and risk of spontaneous preterm birth in Malawian women. iScience 2023; 26:106912. [PMID: 37332611 PMCID: PMC10275952 DOI: 10.1016/j.isci.2023.106912] [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: 06/08/2022] [Revised: 11/11/2022] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
Preterm birth is a leading cause of death in children under five years of age. We hypothesized that sequential disruptions to inflammatory and angiogenic pathways during pregnancy increase the risk of placental insufficiency and spontaneous preterm labor and delivery. We conducted a secondary analysis of inflammatory and angiogenic analytes measured in plasma samples collected across pregnancy from 1462 Malawian women. Women with concentrations of the inflammatory markers sTNFR2, CHI3L1, and IL18BP in the highest quartile before 24 weeks gestation and women with anti-angiogenic factors sEndoglin and sFlt-1/PlGF ratio in the highest quartile at 28-33 weeks gestation had an increased relative risk of preterm birth. Mediation analysis further supported a potential causal link between early inflammation, subsequent angiogenic dysregulation detrimental to placental vascular development, and earlier gestational age at delivery. Interventions designed to reduce the burden of preterm birth may need to be implemented before 24 weeks of gestation.
Collapse
Affiliation(s)
- Andrea M. Weckman
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Robyn E. Elphinstone
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - John M. Ssenkusu
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University, Kampala, Uganda
| | - Vanessa Tran
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Kathleen Zhong
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | | | - Carole Khairallah
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Victor Mwapasa
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Andrea L. Conroy
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Feiko O. Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Chloe R. McDonald
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Kevin C. Kain
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
- Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Research Institute, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| |
Collapse
|
12
|
Shields PG, Ying KL, Brasky TM, Freudenheim JL, Li Z, McElroy JP, Reisinger SA, Song MA, Weng DY, Wewers MD, Whiteman NB, Yang Y, Mathé EA. A Pilot Cross-Sectional Study of Immunological and Microbiome Profiling Reveals Distinct Inflammatory Profiles for Smokers, Electronic Cigarette Users, and Never-Smokers. Microorganisms 2023; 11:1405. [PMID: 37374908 PMCID: PMC10303504 DOI: 10.3390/microorganisms11061405] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Smokers (SM) have increased lung immune cell counts and inflammatory gene expression compared to electronic cigarette (EC) users and never-smokers (NS). The objective of this study is to further assess associations for SM and EC lung microbiomes with immune cell subtypes and inflammatory gene expression in samples obtained by bronchoscopy and bronchoalveolar lavage (n = 28). RNASeq with the CIBERSORT computational algorithm were used to determine immune cell subtypes, along with inflammatory gene expression and microbiome metatranscriptomics. Macrophage subtypes revealed a two-fold increase in M0 (undifferentiated) macrophages for SM and EC users relative to NS, with a concordant decrease in M2 (anti-inflammatory) macrophages. There were 68, 19, and 1 significantly differentially expressed inflammatory genes (DEG) between SM/NS, SM/EC users, and EC users/NS, respectively. CSF-1 and GATA3 expression correlated positively and inversely with M0 and M2 macrophages, respectively. Correlation profiling for DEG showed distinct lung profiles for each participant group. There were three bacteria genera-DEG correlations and three bacteria genera-macrophage subtype correlations. In this pilot study, SM and EC use were associated with an increase in undifferentiated M0 macrophages, but SM differed from EC users and NS for inflammatory gene expression. The data support the hypothesis that SM and EC have toxic lung effects influencing inflammatory responses, but this may not be via changes in the microbiome.
Collapse
Affiliation(s)
- Peter G. Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Department Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Kevin L. Ying
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| | - Theodore M. Brasky
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Department Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Jo L. Freudenheim
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, NY 14261, USA
| | - Zihai Li
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Joseph P. McElroy
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Sarah A. Reisinger
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Min-Ae Song
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Y. Weng
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Mark D. Wewers
- Pulmonary and Critical Care Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Noah B. Whiteman
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Yiping Yang
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Ewy A. Mathé
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institute of Health, Rockville, MD 20892, USA
| |
Collapse
|
13
|
Lee MJ, Kim HS, Kim JH, Choi W, Lee S. Chitinase 3-like 1 induces cancer cell migration in solitary fibrous tumors. Biochem Biophys Res Commun 2023; 663:142-146. [PMID: 37121124 DOI: 10.1016/j.bbrc.2023.04.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/02/2023]
Abstract
Solitary fibrous tumor (SFT) is a rare mesenchymal tumor that is diagnosed through the detection of the NAB2-STAT6 fusion gene. SFT rarely progresses to malignant tumors; however, metastasis is exhibited in approximately 20% of patients with SFT. In this study, we found that chitinase 3-like 1 (CHI3L1), which induces cancer cell migration, was upregulated in NIH-3T3 cells that were transfected with the NAB2-STAT6 fusion gene. Moreover, the expression levels of the migration markers MMP2 and MMP9 were increased and the p-Akt level was also upregulated. In addition, it was observed that when CHI3L1 siRNA was transfected into NAB2-STAT6-transfected cells, cell migration and proliferation were reduced. Therefore, this study demonstrated that CHI3L1 activates Akt signaling to induce cell migration.
Collapse
Affiliation(s)
- Myeong-Ju Lee
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hyeng-Soo Kim
- Institute of Life Science and Biotechnology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ju-Heon Kim
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - WonWoo Choi
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Sanggyu Lee
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
14
|
Shantha Kumara H, Poppy A, Gamage DN, Mitra N, Yan X, Hedjar Y, Cekic V, Whelan RL. Compared to preoperative plasma levels post-operative urokinase-type plasminogen activator-1 levels are persistently elevated for 6 weeks after minimally invasive colorectal resection. J Gastrointest Oncol 2023; 14:187-197. [PMID: 36915462 PMCID: PMC10007942 DOI: 10.21037/jgo-22-113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 10/27/2022] [Indexed: 02/16/2023] Open
Abstract
Background Urokinase-type plasminogen activator-1 (uPA) is a serine protease that converts plasminogen to plasmin after binding to uPA receptor (uPAR). Plasmin catalyzes the regeneration of basement membrane, extracellular matrix, and other tissues. uPA alone and with plasmin leads to activation of angiogenic growth factors that impact tumor cell proliferation, adhesion, and migration. uPA over expression has been noted in colorectal cancer (CRC) and high tissue levels have been correlated with prognosis. uPA/uPAR promotes immune cell activation in healing surgical wounds and may alter perioperative uPA plasma levels. Postoperative (postop) plasma levels, if elevated, may impact the early growth of residual metastases. The impact of minimally invasive colorectal resection (MICR) surgery for CRC on plasma uPA levels is unknown. This study's aim was to measure plasma uPA levels during the first postop month. Methods CRC patients undergoing MICR who enrolled in an Institutional Review Board (IRB) approved data/plasma bank for whom adequate plasma was available were included in the study. Patients who had chemotherapy or radiotherapy within 4 weeks, those who received blood transfusions perioperatively and immunosuppressed patients were excluded. Clinical and pathological data were prospectively collected as were blood samples preoperatively, postop day (POD) 1, 3 and at least 1 late time point between POD 7-41. Plasma was isolated and stored at -80 ℃. Late samples were bundled into 7-day blocks and considered as single time points. Total uPA levels (ng/mL) were analyzed in duplicate via enzyme-linked immunosorbent assay (ELISA) and results reported as mean ± standard deviation (SD). The Wilcoxon paired t-test was used for analysis. Results Ninety-three patients undergoing MICR for CRC [colonic 68%; rectal 32%; average age 65.6 years, laparoscopic 63%, hand-assisted minimally invasive surgery (MIS) 37%] who met criteria were studied. Cancer stage breakdown was; stage I, 30%, stage II, 29%, stage III, 34%, stage IV, 7%. The median preoperative (preop) uPA plasma level (ng/mL) was 529.8 [95% confidence interval (CI): 462.8, 601.1] (n=93). Significant elevations in median levels vs. preop were present during POD 3 (542.8, 95% CI: 518.8, 597.3, n=86, P=0.003), POD 7-13 (688.1, 95% CI: 591.7, 753.0, n=72, P<0.001), POD 14-20 (764.9, 95% CI: 704.1, 911.6, n=27, P<0.001), POD 21-27 (685.6, 95% CI: 443.8, 835.8, n=15, P<0.001), and on POD 28-41 (800.3, 95% CI: 626.9, 940.6, n=21, P<0.001). The colon cancer subgroup's preop and POD 14-20 median results were significantly higher than the corresponding rectal cancer results; otherwise, at the other 5 postop time points there were no significant differences between the rectal and colon cancer subgroups. In addition, no association was found between cancer stage and preop uPA levels and no significant differences were found in postop uPA levels between the hand-assisted laparoscopic group and the lap assisted subgroup at any of the postop time points. Conclusions Persistently elevated plasma uPA levels at 5/6 postop time point (P<0.05), in combination with other previously demonstrated long duration proangiogenic plasma protein changes, may render the plasma proangiogenic within the period of the first month post-surgery and may promote angiogenesis within the residual tumor foci. The clinical significance pertaining to these changes, if any, is uncertain and remains to be proven.
Collapse
Affiliation(s)
- Hmc Shantha Kumara
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Addison Poppy
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Dasuni N Gamage
- Nuvance Health, Vassar Brothers Medical Center, Poughkeepsie, NY, USA
| | - Neil Mitra
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Xiaohong Yan
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Yanni Hedjar
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Vesna Cekic
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Richard L Whelan
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell 500 Hofstra Blvd., Hempstead, NY, USA
| |
Collapse
|
15
|
Bader JM, Deigendesch N, Misch M, Mann M, Koch A, Meissner F. Proteomics separates adult-type diffuse high-grade gliomas in metabolic subgroups independent of 1p/19q codeletion and across IDH mutational status. Cell Rep Med 2023; 4:100877. [PMID: 36584682 PMCID: PMC9873829 DOI: 10.1016/j.xcrm.2022.100877] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 07/15/2022] [Accepted: 12/07/2022] [Indexed: 12/30/2022]
Abstract
High-grade adult-type diffuse gliomas are malignant neuroepithelial tumors with poor survival rates in combined chemoradiotherapy. The current WHO classification is based on IDH1/2 mutational and 1p/19q codeletion status. Glioma proteome alterations remain undercharacterized despite their promise for a better molecular patient stratification and therapeutic target identification. Here, we use mass spectrometry to characterize 42 formalin-fixed, paraffin-embedded (FFPE) samples from IDH-wild-type (IDHwt) gliomas, IDH-mutant (IDHmut) gliomas with and without 1p/19q codeletion, and non-neoplastic controls. Based on more than 5,500 quantified proteins and 5,000 phosphosites, gliomas separate by IDH1/2 mutational status but not by 1p/19q status. Instead, IDHmut gliomas split into two proteomic subtypes with widespread perturbations, including aerobic/anaerobic energy metabolism. Validations with three independent glioma proteome datasets confirm these subgroups and link the IDHmut subtypes to the established proneural and classic/mesenchymal subtypes in IDHwt glioma. This demonstrates common phenotypic subtypes across the IDH status with potential therapeutic implications for patients with IDHmut gliomas.
Collapse
Affiliation(s)
- Jakob Maximilian Bader
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Nikolaus Deigendesch
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Martin Misch
- Department of Neurosurgery, Charité, Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Arend Koch
- Department of Neuropathology, Charité, Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany.
| | - Felix Meissner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Department of Systems Immunology and Proteomics, Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany.
| |
Collapse
|
16
|
Ma Z, Wang G, Hu L. lncRNA EGFEM1P Drives the Progression of Papillary Thyroid Cancer by Regulating miR-6867-5p/CHI3L1 Axis. Crit Rev Eukaryot Gene Expr 2023; 33:29-41. [PMID: 37522543 DOI: 10.1615/critreveukaryotgeneexpr.2023047995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Long noncoding RNA (lncRNA), a subgroup of noncoding RNA with > 200 nt, plays critical roles in cancer progression. Here, we aimed to explore the detailed biological function of lncRNA EGFEM1P during papillary thyroid cancer (PTC) progression. RT-qPCR and Western blot were used to analyze the expression of lncRNA EGFEM1P, miR-6867-5p, and CHI3L1. CCK8, colony formation, and Transwell migration assays were undertaken to assess PTC cell proliferation and migration. A xenograft tumor mouse model was also used to establish tumor growth in vivo. Luciferase reporter and anti-AGO2 RNA immunoprecipitation (RIP) assays were used to clarify the interplay between miR-6867-5p and lncRNA EGFEM1P or CHI3L1. We found lncRNA EGFEM1P and CHI3L1 to be highly expressed in PTC tissues and cells, while miR-6867-5p expression decreases. Functionally, lncRNA EGFEM1P silence delays PTC cell proliferation and migration, and impairs tumorigenesis in vivo. LncRNA EGFEM1P targets miR-6867-5p, and CHI3L1 is a target gene of miR-6867-5p. LncRNA EGFEM1P silence decreases the pro-proliferation and pro-migration caused by the miR-6867-5p inhibitor in PTC cells, and CHI3L1 silence abrogates the pro-tumorigenic action resulting from the miR-6867-5p inhibitor in PTC cells. Our data showed that lncRNA EGFEM1P targeting of the miR-6867-5p/CHI3L1 axis drives PTC progression, suggesting lncRNA EGFEM1P as a therapeutically target for PTC.
Collapse
Affiliation(s)
- Zhanwu Ma
- Department of Medical Clinical Laboratory, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Guoxian Wang
- Department of Radiology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Lin Hu
- Department of Medical Clinical Laboratory, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, Hubei, China
| |
Collapse
|
17
|
Chen WW, Chu TSM, Xu L, Zhao CN, Poon WS, Leung GKK, Kong FMS. Immune related biomarkers for cancer metastasis to the brain. Exp Hematol Oncol 2022; 11:105. [PMID: 36527157 PMCID: PMC9756766 DOI: 10.1186/s40164-022-00349-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/14/2022] [Indexed: 12/23/2022] Open
Abstract
Brain metastasis accounts for a large number of cancer-related deaths. The host immune system, involved at each step of the metastatic cascade, plays an important role in both the initiation of the brain metastasis and their treatment responses to various modalities, through either local and or systemic effect. However, few reliable immune biomarkers have been identified in predicting the development and the treatment outcome in patients with cancer brain metastasis. Here, we provide a focused perspective of immune related biomarkers for cancer metastasis to the brain and a thorough discussion of the potential utilization of specific biomarkers such as tumor mutation burden (TMB), genetic markers, circulating and tumor-infiltrating immune cells, cytokines, in predicting the brain disease progression and regression after therapeutic intervention. We hope to inspire the field to extend the research and establish practical guidelines for developing and validating immune related biomarkers to provide personalized treatment and improve treatment outcomes in patients with metastatic brain cancers.
Collapse
Affiliation(s)
- Wei-Wei Chen
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, SAR, China
| | - Timothy Shun Man Chu
- Royal Victoria Infirmary, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Queen Victoria Road, Newcastle Upon Tyne, NE1 4LP, UK
- Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - LiangLiang Xu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Cai-Ning Zhao
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, SAR, China
| | - Wai-Sang Poon
- Neuro-Medical Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Surgery, School of Clinical Medicine,LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, SAR, China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, School of Clinical Medicine,LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, SAR, China
| | - Feng-Ming Spring Kong
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, SAR, China.
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
18
|
Robilliard LD, Yu J, Anchan A, Finlay G, Angel CE, Graham ES. Comprehensive Assessment of Secreted Immuno-Modulatory Cytokines by Serum-Differentiated and Stem-like Glioblastoma Cells Reveals Distinct Differences between Glioblastoma Phenotypes. Int J Mol Sci 2022; 23:ijms232214164. [PMID: 36430641 PMCID: PMC9692434 DOI: 10.3390/ijms232214164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/04/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma is refractory to therapy and presents a significant oncological challenge. Promising immunotherapies have not shown the promise observed in other aggressive cancers. The reasons for this include the highly immuno-suppressive tumour microenvironment controlled by the glioblastoma cells and heterogeneous phenotype of the glioblastoma cells. Here, we wanted to better understand which glioblastoma phenotypes produced the regulatory cytokines, particularly those that are implicated in shaping the immune microenvironment. In this study, we employed nanoString analysis of the glioblastoma transcriptome, and proteomic analysis (proteome profiler arrays and cytokine profiling) of secreted cytokines by different glioblastoma phenotypes. These phenotypes were cultured to reflect a spectrum of glioblastoma cells present in tumours, by culturing an enhanced stem-like phenotype of glioblastoma cells or a more differentiated phenotype following culture with serum. Extensive secretome profiling reveals that there is considerable heterogeneity in secretion patterns between serum-derived and glioblastoma stem-like cells, as well as between individuals. Generally, however, the serum-derived phenotypes appear to be the primary producers of cytokines associated with immune cell recruitment into the tumour microenvironment. Therefore, these glioblastoma cells have considerable importance in shaping the immune landscape in glioblastoma and represent a valuable therapeutic target that should not be ignored.
Collapse
Affiliation(s)
- Laverne D. Robilliard
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand
| | - Jane Yu
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand
| | - Akshata Anchan
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand
| | - Graeme Finlay
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Auckland Cancer Society Research Centre, University of Auckland, Auckland 1023, New Zealand
| | - Catherine E. Angel
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1010, New Zealand
| | - E Scott Graham
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand
- Correspondence:
| |
Collapse
|
19
|
Park YJ, Rahman MS, Pang WK, Ryu DY, Jung MJ, Amjad S, Kim JM, Pang MG. Systematic multi-omics reveals the overactivation of T cell receptor signaling in immune system following bisphenol A exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 308:119590. [PMID: 35752395 DOI: 10.1016/j.envpol.2022.119590] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/17/2022] [Accepted: 06/04/2022] [Indexed: 06/15/2023]
Abstract
Bisphenol A (BPA) is pervasive in the environment, and exposure to BPA may increase the incidence of noncommunicable diseases like autoimmune diseases and cancer. Although BPA causes immunological problems at the cellular level, no system-level research has been conducted on this. Hence, in this study, we aimed to gain a better understanding of the biological response to BPA exposure and its association with immunological disorders. For that, we explored the transcriptome and the proteomic modifications at the systems and cellular levels following BPA exposure. Our integrated multi-omics data showed the alteration of the T cell receptor (TCR) signaling pathway at both levels. The proportion of enlarged T cells increased with upregulation of CD69, a surface marker of early T cell activation, even though the number of T cells reduced after BPA exposure. Additionally, on BPA exposure, the levels of pLCK and pSRC increased in T cells, while that of pLAT decreased. Following BPA exposure, we investigated cytokine profiles and discovered that chitinase 3 Like 1 and matrix metalloproteinase 9 were enriched in T cells. These results indicated that T cells were hyperactivated by CD69 stimulation, and phosphorylation of SRC accelerated on BPA exposure. Hence, alteration in the TCR signaling pathway during development and differentiation due to BPA exposure could lead to insufficient and hasty activation of TCR signaling in T cells, which could modify cytokine profiles, leading to increased environmental susceptibility to chronic inflammation or diseases, increasing the chance of autoimmune diseases and cancer. This study enhances our understanding of the effects of environmental perturbations on immunosuppression at molecular, cellular, and systematic levels following pubertal BPA exposure, and may help develop better predictive, preventative, and therapeutic techniques.
Collapse
Affiliation(s)
- Yoo-Jin Park
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, South Korea
| | - Md Saidur Rahman
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, South Korea
| | - Won-Ki Pang
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, South Korea
| | - Do-Yeal Ryu
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, South Korea
| | - Min-Ji Jung
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, South Korea
| | - Shehreen Amjad
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, South Korea
| | - Jun-Mo Kim
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, South Korea
| | - Myung-Geol Pang
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, South Korea.
| |
Collapse
|
20
|
Hegde M, Daimary UD, Kumar A, Chinnathambi A, Alharbi SA, Shakibaei M, Kunnumakkara AB. STAT3/HIF1A and EMT specific transcription factors regulated genes: Novel predictors of breast cancer metastasis. Gene X 2022; 818:146245. [PMID: 35074419 DOI: 10.1016/j.gene.2022.146245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/18/2022] [Indexed: 12/26/2022] Open
Abstract
Metastasis, the fatal hallmark of breast cancer (BC), is a serious hurdle for therapy. Current prognostic approaches are not sufficient to predict the metastasis risk for BC patients. Therefore, in the present study, we analyzed gene expression data from GSE139038 and TCGA database to develop predictive markers for BC metastasis. Initially, the data from GSE139038 which contained 65 samples consisting of 41 breast tumor tissues, 18 paired morphologically normal tissues and 6 from non-malignant breast tissues were analyzed for differentially expressed genes (DEGs). DEGs were obtained from three different comparisons: paired morphologically normal (MN) versus tumor samples (C), apparently normal (AN) versus tumor samples (C), and paired morphologically normal (MN) versus apparently normal samples (AN). Multiple bioinformatic methods were employed to evaluate metastasis, EMT and triple negative breast cancer (TNBC) specific genes. Further, regulation of gene expression, clinicopathological factors and DNA methylation patterns of DEGs in BC were validated with TCGA datasets. Our bioinformatic analysis showed that 40 genes were upregulated and 294 were found to be downregulated between AN vs C; 124 were upregulated and 760 genes were downregulated between MN vs C; 4 were upregulated and 13 were downregulated between MN vs AN. Analysis using TCGA dataset revealed 18 genes were significantly altered in nodal positive BC patients compared to nodal negative BC patients. Our study showed novel candidate genes as predictive markers for BC metastasis which can also be used for therapeutic targets for BC treatment.
Collapse
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781 039, Assam, India; DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati 781 039, Assam, India
| | - Uzini Devi Daimary
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781 039, Assam, India; DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati 781 039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781 039, Assam, India; DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati 781 039, Assam, India
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box 2455, Riyadh 11451, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box 2455, Riyadh 11451, Saudi Arabia
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781 039, Assam, India; DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati 781 039, Assam, India.
| |
Collapse
|
21
|
Luo L, Santos A, Konganti K, Hillhouse A, Lambertz IU, Zheng Y, Gunaratna RT, Threadgill DW, Fuchs-Young RS. Overexpression of IGF-1 During Early Development Expands the Number of Mammary Stem Cells and Primes them for Transformation. Stem Cells 2022; 40:273-289. [DOI: 10.1093/stmcls/sxab018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Indexed: 11/13/2022]
Abstract
Abstract
Insulin-like growth factor I (IGF-1) has been implicated in breast cancer due to its mitogenic and anti-apoptotic effects. Despite substantial research on the role of IGF-1 in tumor progression, the relationship of IGF-1 to tissue stem cells, particularly in mammary tissue, and the resulting tumor susceptibility has not been elucidated. Previous studies with the BK5.IGF-1 transgenic (Tg) mouse model reveals that IGF-1 does not act as a classical, post-carcinogen tumor promoter in the mammary gland. Pre-pubertal Tg mammary glands display increased numbers and enlarged sizes of terminal end buds, a niche for mammary stem cells (MaSCs). Here we show that MaSCs from both wild type (WT) and Tg mice expressed IGF-1R and that overexpression of Tg IGF-1 increased numbers of MaSCs by undergoing symmetric division, resulting in an expansion of the MaSC and luminal progenitor (LP) compartments in pre-pubertal female mice. This expansion was maintained post-pubertally and validated by mammosphere assays in vitro and transplantation assays in vivo. The addition of recombinant IGF-1 promoted, and IGF-1R downstream inhibitors decreased mammosphere formation. Single-cell transcriptomic profiles generated from two related platforms reveal that IGF-1 stimulated quiescent MaSCs to enter the cell cycle and increased their expression of genes involved in proliferation, plasticity, tumorigenesis, invasion, and metastasis. This study identifies a novel, pro-tumorigenic mechanism, where IGF-1 increases the number of transformation-susceptible carcinogen targets during the early stages of mammary tissue development, and “primes” their gene expression profiles for transformation.
Collapse
Affiliation(s)
- Linjie Luo
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
- Department of Experimental Radiation Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andres Santos
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
- Department of Anatomic Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kranti Konganti
- Texas A&M Institute for Genome Sciences & Society, Texas A&M University, College Station, TX, USA
| | - Andrew Hillhouse
- Texas A&M Institute for Genome Sciences & Society, Texas A&M University, College Station, TX, USA
| | - Isabel U Lambertz
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Yuanning Zheng
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Ramesh T Gunaratna
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - David W Threadgill
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
- Texas A&M Institute for Genome Sciences & Society, Texas A&M University, College Station, TX, USA
| | - Robin S Fuchs-Young
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| |
Collapse
|
22
|
Groysman L, Carlsen L, Huntington KE, Shen WH, Zhou L, El-Deiry WS. Chemotherapy-induced cytokines and prognostic gene signatures vary across breast and colorectal cancer. Am J Cancer Res 2021; 11:6086-6106. [PMID: 35018244 PMCID: PMC8727797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/12/2021] [Indexed: 06/14/2023] Open
Abstract
The mechanisms by which chemotherapeutic drugs mediate efficacy and toxicity in patients across cancers are not fully understood. A poorly understood aspect of the tumor cell response to chemotherapy is cytokine regulation. Some drug-induced cytokines promote the anti-cancer activity of the drugs, but others may promote proliferation, metastasis, and drug resistance. We evaluated effects of clinical chemotherapeutics oxaliplatin, cisplatin, 5-fluorouracil (5-FU), doxorubicin, paclitaxel, docetaxel, and carboplatin on a panel of 52 cytokines in MCF7 breast cancer (BC) cells. We observed pan-drug effects, such as the upregulation of TRAIL-R2 and Chitinase 3-like 1 and drug-specific effects on interleukin and CXCL cytokines. We compared cytokine regulation in MCF7 BC and HCT116 colorectal cancer (CRC) cells, revealing tissue-specific drug effects such as enhanced upregulation of TRAIL-R2 and downregulation of IFN-β and TRAIL in MCF7 by cisplatin, oxaliplatin, and 5-FU. We found that chemotherapy-inducible transcripts have varying potential for prognostic significance in CRC versus BC. Among the non-prognostic CRC genes that were prognostic in BC were NFKBIA and GADD45A, both of which support anti-cancer drug mechanisms. Thus, we establish a novel 7-drug, 52-cytokine signature in MCF7 BC cells and a 3-drug, 40-cytokine signature in HCT116 CRC cells that suggest drug-specific and tissue-specific cytokine regulation. Distinct differences across prognostic gene signatures in BC and CRC further support tissue specificity in the relative impact of drug-regulated genes on patient survival.
Collapse
Affiliation(s)
- Leya Groysman
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Macaulay Honors College at Hunter College, CUNYManhattan, NY 10065, USA
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell UniversityNY 10065, USA
| | - Lindsey Carlsen
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Graduate Program in Pathobiology, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Kelsey E Huntington
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Graduate Program in Pathobiology, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Wen H Shen
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell UniversityNY 10065, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Graduate Program in Pathobiology, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Hematology-Oncology Division, Brown University and The Lifespan Cancer InstituteProvidence, RI 02903, USA
- Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| |
Collapse
|
23
|
Shantha Kumara HMC, Shah A, Miyagaki H, Yan X, Cekic V, Hedjar Y, Whelan RL. Plasma Levels of Keratinocyte Growth Factor Are Significantly Elevated for 5 Weeks After Minimally Invasive Colorectal Resection Which May Promote Cancer Recurrence and Metastasis. Front Surg 2021; 8:745875. [PMID: 34820416 PMCID: PMC8606552 DOI: 10.3389/fsurg.2021.745875] [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: 07/22/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Human Keratinocyte Growth Factor (KGF) is an FGF family protein produced by mesenchymal cells. KGF promotes epithelial cell proliferation, plays a role in wound healing and may also support tumor growth. It is expressed by some colorectal cancers (CRC). Surgery's impact on KGF levels is unknown. This study's purpose was to assess plasma KGF levels before and after minimally invasive colorectal resection (MICR) for CRC. Aim: To determine plasma KGF levels before and after minimally invasive colorectal resection surgery for cancer pathology. Method: CRC MICR patients (pts) in an IRB approved data/plasma bank were studied. Pre-operative (pre-op) and post-operative (post-op) plasma samples were taken/stored. Late samples were bundled into 7 day blocks and considered as single time points. KGF levels (pg/ml) were measured via ELISA (mean ± SD). The Wilcoxon paired t-test was used for statistical analysis. Results: Eighty MICR CRC patients (colon 61%; rectal 39%; mean age 65.8 ± 13.3) were studied. The mean incision length was 8.37 ± 3.9 and mean LOS 6.5 ± 2.6 days. The cancer stage breakdown was; I (23), II (26), III (27), and IV (4). The median pre-op KGF level was 17.1 (95 %CI: 14.6-19.4; n = 80); significantly elevated (p < 0.05) median levels (pg/ml) were noted on post-op day (POD) 1 (23.4 pg/ml; 95% CI: 21.4-25.9; n = 80), POD 3 (22.5 pg/ml; 95% CI: 20.7-25.9; n = 76), POD 7-13 (21.8 pg/ml; 95% CI: 17.7-25.4; n = 50), POD 14-20 (20.1 pg/ml; 95% CI: 17.1-23.9; n = 33), POD 21-27 (19.6 pg/ml; 95% CI: 15.2-24.9; n = 15) and on POD 28-34 (16.7 pg/ml; 95% CI: 14.0-25.8; n = 12). Conclusion: Plasma KGF levels were significantly elevated for 5 weeks after MICR for CRC. The etiology of these changes is unclear, surgical trauma related acute inflammatory response and wound healing process may play a role. These changes, may stimulate angiogenesis in residual tumor deposits after surgery.
Collapse
Affiliation(s)
- H M C Shantha Kumara
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Abhinit Shah
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | | | - Xiaohong Yan
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Vesna Cekic
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Yanni Hedjar
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Richard L Whelan
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
24
|
Williams MM, Hafeez SA, Christenson JL, O’Neill KI, Hammond NG, Richer JK. Reversing an Oncogenic Epithelial-to-Mesenchymal Transition Program in Breast Cancer Reveals Actionable Immune Suppressive Pathways. Pharmaceuticals (Basel) 2021; 14:ph14111122. [PMID: 34832904 PMCID: PMC8622696 DOI: 10.3390/ph14111122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
Approval of checkpoint inhibitors for treatment of metastatic triple negative breast cancer (mTNBC) has opened the door for the use of immunotherapies against this disease. However, not all patients with mTNBC respond to current immunotherapy approaches such as checkpoint inhibitors. Recent evidence demonstrates that TNBC metastases are more immune suppressed than primary tumors, suggesting that combination or additional immunotherapy strategies may be required to activate an anti-tumor immune attack at metastatic sites. To identify other immune suppressive mechanisms utilized by mTNBC, our group and others manipulated oncogenic epithelial-to-mesenchymal transition (EMT) programs in TNBC models to reveal differences between this breast cancer subtype and its more epithelial counterpart. This review will discuss how EMT modulation revealed several mechanisms, including tumor cell metabolism, cytokine milieu and secretion of additional immune modulators, by which mTNBC cells may suppress both the innate and adaptive anti-tumor immune responses. Many of these pathways/proteins are under preclinical or clinical investigation as therapeutic targets in mTNBC and other advanced cancers to enhance their response to chemotherapy and/or checkpoint inhibitors.
Collapse
|
25
|
Oh IH, Pyo JS, Son BK. Prognostic Impact of YKL-40 Immunohistochemical Expression in Patients with Colorectal Cancer. Curr Oncol 2021; 28:3139-3149. [PMID: 34436040 PMCID: PMC8395453 DOI: 10.3390/curroncol28040274] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022] Open
Abstract
This study aims to examine the clinicopathological and prognostic significance of the YKL-40 immunohistochemical expression of tumor and immune cells through human colorectal cancer (CRC) tissue. We performed immunohistochemistry for YKL-40 and investigated the clinicopathological and prognostic impact of the YKL-40 expression of tumor (T-YKL-40) and immune cells (I-YKL-40) in CRC. We also evaluated the correlation between YKL-40 and PD-L1 expression and the immunoscore. YKL-40 was expressed in 22.6% and 64.2% of T-YKL-40 and I-YKL-40, respectively, out of the 265 CRC tissues. The I-YKL-40 expression significantly correlated with well and moderately differentiated tumors. The PD-L1 expression in immune cells significantly correlated with the I-YKL-40 expression, but not T-YKL-40 expression (p = 0.020 and p = 0.846, respectively). The I-YKL-40 expression significantly correlated with a worse overall survival rate but not recurrence-free survival (p = 0.047 and p = 0.080, respectively). However, there was no significant correlation between the T-YKL-40 expression and survival. In CRCs with a high immunoscore, patients with I-YKL-40 expression demonstrated worse overall and recurrence-free survival than those without I-YKL-40 expression. Our results demonstrated that I-YKL-40 expression significantly correlated with tumor differentiation and PD-L1 expression in immune cells. I-YKL-40 expression can be useful for the prognostic stratification of CRC patients.
Collapse
Affiliation(s)
- Il Hwan Oh
- Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu-si 11759, Korea;
| | - Jung-Soo Pyo
- Department of Pathology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu-si 11759, Korea;
| | - Byoung Kwan Son
- Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu-si 11759, Korea;
| |
Collapse
|
26
|
Xue Q, Chen L, Yu J, Sun K, Ye L, Zheng J. Downregulation of Interleukin-13 Receptor α2 Inhibits Angiogenic Formation Mediated by Chitinase 3-Like 1 in Late Atherosclerotic Lesions of apoE -/- Mice. Front Physiol 2021; 12:690109. [PMID: 34349665 PMCID: PMC8327173 DOI: 10.3389/fphys.2021.690109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/03/2021] [Indexed: 11/13/2022] Open
Abstract
Aim: Chitinase 3-like 1 (CHI3L1) has the potential to prompt proliferation and angiogenic formation. Interleukin-13 receptor α2 (IL-13Rα2) was regarded as a receptor of CHI3L1; however, it is unknown whether CHI3L1 adjusts the neovascularization in late atherosclerotic lesions of apoE -/- mice via IL-13Rα2. Methods: Silicone collars were placed around one of the common carotid arteries of apoE -/- mice fed with a high-fat diet. The mice were further injected with Ad.CHI3L1 alone or Ad.CHI3L1 + Ad.IL-13Rα2 shRNA through the caudal vein. The plaque areas in the whole aorta and aortic root were evaluated by Oil Red O staining and H&E staining. The contents of CD31, CD42b, and collagen in carotid plaques were investigated by immunohistochemistry and Masson trichrome staining. The role of CHI3L1 in migration and tube formation of human umbilical vein endothelial cells (HUVECs) was determined by transwell and Matrigel tests. The effect of CHI3L1 on the expression of AKT and extracellular signal-regulated kinase (ERK) was evaluated with the Western blot. Results: The plaque loads in the aorta were significantly more extensive in apoE -/- mice injected with Ad.CHI3L1 than those with Ad.CHI3L1 + Ad.IL-13Rα2 shRNA. CHI3L1 significantly increased the contents of CD31 and CD42b and decreased the element of collagen in late-stage atherosclerotic lesions of the carotid arteries. The effects of CHI3L1 on migration, tube formation, and upregulation of phospho-AKT and phospho-ERK of HUVECs were prohibited by inhibitors of phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase kinase (MEK) as well as IL-13Rα2 shRNA. Conclusion: To some extent, CHI3L1 promotes migration and tube formation of HUVECs and neovascularization in atherosclerotic plaques possibly mediated by IL-13Rα2 through AKT and ERK signal pathways.
Collapse
Affiliation(s)
- Qi Xue
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lei Chen
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jianwu Yu
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Kewang Sun
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lifang Ye
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jianlei Zheng
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
27
|
Das A, Kamrul-Hasan ABM, Kabir MR, Das S, Zaki KMJ, Al Mahtab M. Evaluation of Chitinase 3-like 1 (CHI3L1) as a noninvasive biomarker of hepatic fibrosis in patients with Hepatitis B virus-related compensated chronic liver disease. J Family Med Prim Care 2021; 10:1694-1698. [PMID: 34123914 PMCID: PMC8144771 DOI: 10.4103/jfmpc.jfmpc_1922_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/24/2020] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Liver biopsy is the gold-standard method for diagnosing and staging liver fibrosis, but the procedure is invasive, not available in the primary health care facilities, and not free from complications. Noninvasive serum biomarkers of hepatic fibrosis are the current research focus. OBJECTIVES To assess the correlation between serum Chitinase 3-like 1 (CHI3L1) levels and histological severity in patients with Hepatitis B Virus (HBV)-related compensated chronic liver disease (CLD). MATERIAL AND METHODS This cross-sectional study evaluated 50 treatment-naïve patients with chronic hepatitis B with compensated CLD. Liver biopsy was done, and hepatic fibrosis was categorized using the METAVIR scoring system; we divided the study subjects into three groups; group 1 included subjects with F0 and F1, group 2 having F2 group 3 having F3 and F4. Serum CHI3L1 was measured in all by immunoassay. RESULT Among 50 patients, only one had METAVIR score F0, seven had F1, 33 had F2, nine had F3, and none had METAVIR score F4. The median value of CHI3L1 was 460.8 (IQR 340.1-570.3) in all study subjects; 359.5 (IQR 272.8-526.9) in group 1, 450.0 (IQR 307.75-5332.0) in group 2, and 1355.5 (IQR 530.75-1580.5) in the group 3. The difference in median CHI3L1 across the groups was statistically significant. Serum aspartate aminotransferase (AST) and the AST to Platelet Ratio Index (APRI) score had significant positive correlations with CHI3L1 levels. CHI3L1 also had significant positive correlations with METAVIR scores. CONCLUSION This study found a positive correlation between serum CHI3L1 level and hepatic histological severity in patients with HBV-related compensated CLD. Further larger-scale research is needed to establish the fact.
Collapse
Affiliation(s)
- Amit Das
- Department of Medicine, Sylhet MAG Osmani Medical College Hospital, Sylhet, Bangladesh
| | - ABM Kamrul-Hasan
- Department of Endocrinology, Mymensingh Medical College, Mymensingh, Bangladesh
| | - Mohammed Ruhul Kabir
- Department of Medicine, Sylhet MAG Osmani Medical College Hospital, Sylhet, Bangladesh
| | - Shantanu Das
- Department of Microbiology, Sylhet MAG Osmani Medical College Hospital, Sylhet, Bangladesh
| | - KMJ Zaki
- Department of Hepatology, Sylhet MAG Osmani Medical College, Sylhet, Bangladesh
| | - Mamun Al Mahtab
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| |
Collapse
|
28
|
Yang X, Fang D, Li M, Chen J, Cheng Y, Luo J. Knockdown of Chitinase 3-Like-1 Inhibits Cell Proliferation, Promotes Apoptosis, and Enhances Effect of Anti-Programmed Death Ligand 1 (PD-L1) in Diffuse Large B Cell Lymphoma Cells. Med Sci Monit 2021; 27:e929431. [PMID: 33764958 PMCID: PMC8008972 DOI: 10.12659/msm.929431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Enzymatically inactive chitinase-like protein CHI3L1 is overexpressed in diffuse large B cell lymphoma (DLBCL) patients with PD-L1 imbalance and promotes tumor progression in the microenvironment. Based on this, we investigated how CHI3L1 acts on the proliferation and apoptosis of DLBCL and whether there is a synergy of CHI3L1 in combination with anti-PD-L1 antibodies in vivo. MATERIAL AND METHODS CHI3L1 was detected by quantitative real-time PCR (RT-PCR) and western blot (WB) in B-lymphoma cell lines. CHI3L1 interference plasmids were constructed, and the levels of proliferation, cell cycle, apoptosis, and cell survival were examined in vitro in B-lymphoma cell lines and in vivo in a murine xenograft model by RT-PCR, WB, CCK-8, and flow cytometry. RESULTS CHI3L1 was significantly expressed in SU-DHL-4 cells. CHI3L1-interfered RNA ShRNA-CHI3L1-1 was chosen to be used in the next experiment because it had a better interference effect. Dampened cell proliferation level, arrested cell cycle, reduced protein expressions of cyclin D1 and cyclin D2, and promoted cell apoptosis level were observed after SU-DHL-4 was transfected with ShRNA-CHI3L1-1. Furthermore, we also noticed increased expression of Bcl-2, decreased expressions of bax, cleaved caspase 3 and cleaved PARP, promoted cell survival-related protein p53, and reduced survivin. CONCLUSIONS This study demonstrated that knockdown of CHI3L1 inhibits cancer cell proliferation by regulating cell cycles, promotes cancer cell apoptosis, and enhances the pro-apoptotic effect of anti-PD-L1 antibody both in vivo and in vitro in DLBCL.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Dong Fang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Ming Li
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jiayi Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yuanbo Cheng
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jianming Luo
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
29
|
Han JS, Kim SE, Jin JQ, Park NR, Lee JY, Kim HL, Lee SB, Yang SW, Lim DJ. Tear-Derived Exosome Proteins Are Increased in Patients with Thyroid Eye Disease. Int J Mol Sci 2021; 22:ijms22031115. [PMID: 33498689 PMCID: PMC7866068 DOI: 10.3390/ijms22031115] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/14/2022] Open
Abstract
Exosomes contain proteins, lipids, RNA, and DNA that mediate intercellular signaling. Exosomes can contribute to the pathological processes of various diseases, although their roles in ocular diseases are unclear. We aimed to isolate exosomes from tear fluids (TF) of patients with Thyroid eye disease (TED) and analyze the exosomal proteins. TFs were collected from eight patients with TED and eight control subjects. The number of TF exosomes were measured using nanoparticle-tracking analysis. The expression of specific proteins in the purified exosome pellets were analyzed using a Proteome Profiler Array Kit. Cultured normal orbital fibroblasts were incubated with TF exosomes from patients with TED and control subjects, and changes in inflammatory cytokine levels were compared. TF exosomes from TED patients showed more exosomes than the control subjects. The expression levels of exosomal proteins vitamin D-binding (VDB) protein, C-reactive protein (CRP), chitinase 3-like 1 (CHI3L1), matrix metalloproteinase-9 (MMP-9), and vascular adhesion molecule-1 (VCAM-1) were significantly increased in patients with TED, compared to those of controls. Orbital fibroblasts exposed to TF exosomes from patients with TED showed significantly higher levels of interleukin (IL)-6, IL-8, and monocyte chemoattractant protein-1 (MCP-1) production than those treated with control TF exosomes. Specific proteins showed higher expression in exosomes from TED patients, implying that they may play keys roles in TED pathogenesis.
Collapse
Affiliation(s)
- Jeong-Sun Han
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Seoul St. Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (J.-S.H.); (J.-Q.J.)
| | - Sung Eun Kim
- Department of Ophthalmology and Visual Science, Seoul St. Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.E.K.); (N.R.P.)
| | - Jun-Qing Jin
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Seoul St. Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (J.-S.H.); (J.-Q.J.)
| | - Na Ri Park
- Department of Ophthalmology and Visual Science, Seoul St. Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.E.K.); (N.R.P.)
| | - Ji-Young Lee
- Department of Pathology, Institute of Hansen’s Disease, Seoul St. Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (J.-Y.L.); (S.-B.L.)
| | - Hong Lim Kim
- Integrative Research Support Center, Laboratory of Electron Microscope, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Seong-Beom Lee
- Department of Pathology, Institute of Hansen’s Disease, Seoul St. Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (J.-Y.L.); (S.-B.L.)
| | - Suk-Woo Yang
- Department of Ophthalmology and Visual Science, Seoul St. Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.E.K.); (N.R.P.)
- Correspondence: (S.-W.Y.); (D.-J.L.); Tel.: +82-2-2258-6009 (D.-J.L.)
| | - Dong-Jun Lim
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Seoul St. Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (J.-S.H.); (J.-Q.J.)
- Correspondence: (S.-W.Y.); (D.-J.L.); Tel.: +82-2-2258-6009 (D.-J.L.)
| |
Collapse
|
30
|
Satitsri S, Muanprasat C. Chitin and Chitosan Derivatives as Biomaterial Resources for Biological and Biomedical Applications. Molecules 2020; 25:molecules25245961. [PMID: 33339290 PMCID: PMC7766609 DOI: 10.3390/molecules25245961] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 01/30/2023] Open
Abstract
Chitin is a long-chain polymer of N-acetyl-glucosamine, which is regularly found in the exoskeleton of arthropods including insects, shellfish and the cell wall of fungi. It has been known that chitin can be used for biological and biomedical applications, especially as a biomaterial for tissue repairing, encapsulating drug for drug delivery. However, chitin has been postulated as an inducer of proinflammatory cytokines and certain diseases including asthma. Likewise, chitosan, a long-chain polymer of N-acetyl-glucosamine and d-glucosamine derived from chitin deacetylation, and chitosan oligosaccharide, a short chain polymer, have been known for their potential therapeutic effects, including anti-inflammatory, antioxidant, antidiarrheal, and anti-Alzheimer effects. This review summarizes potential utilization and limitation of chitin, chitosan and chitosan oligosaccharide in a variety of diseases. Furthermore, future direction of research and development of chitin, chitosan, and chitosan oligosaccharide for biomedical applications is discussed.
Collapse
|
31
|
Ham HJ, Lee YS, Yun J, Son DJ, Lee HP, Han SB, Hong JT. K284-6111 alleviates memory impairment and neuroinflammation in Tg2576 mice by inhibition of Chitinase-3-like 1 regulating ERK-dependent PTX3 pathway. J Neuroinflammation 2020; 17:350. [PMID: 33222690 PMCID: PMC7681957 DOI: 10.1186/s12974-020-02022-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Background Alzheimer’s disease (AD) is one of the most prevalent neurodegenerative disorders characterized by gradual memory loss and neuropsychiatric symptoms. We have previously demonstrated that the 2-({3-[2-(1-cyclohexene-1-yl)ethyl]-6,7-dimethoxy-4-oxo-3,4-dihydro-2-quinazolinyl}sulfanyl)-N-(4-ethylphenyl)butanamide (K284-6111), the inhibitor of CHI3L1, has the inhibitory effect on memory impairment in Αβ infusion mouse model and on LPS-induced neuroinflammation in the murine BV-2 microglia and primary cultured astrocyte. Methods In the present study, we investigated the inhibitory effect of K284-6111 on memory dysfunction and neuroinflammation in Tg2576 transgenic mice, and a more detailed correlation of CHI3L1 and AD. To investigate the effects of K284-6111 on memory dysfunction, we administered K284-6111 (3 mg/kg, p.o.) daily for 4 weeks to Tg2576 mice, followed by behavioral tests of water maze test, probe test, and passive avoidance test. Results Administration of K284-6111 alleviated memory impairment in Tg2576 mice and had the effect of reducing the accumulation of Aβ and neuroinflammatory responses in the mouse brain. K284-6111 treatment also selectively inactivated ERK and NF-κB pathways, which were activated when CHI3L1 was overexpressed, in the mouse brain and in BV-2 cells. Web-based gene network analysis and our results of gene expression level in BV-2 cells showed that CHI3L1 is closely correlated with PTX3. Our result revealed that knockdown of PTX3 has an inhibitory effect on the production of inflammatory proteins and cytokines, and on the phosphorylation of ERK and IκBα. Conclusion These results suggest that K284-6111 could improve memory dysfunction by alleviating neuroinflammation through inhibiting CHI3L1 enhancing ERK-dependent PTX3 pathway.
Collapse
Affiliation(s)
- Hyeon Joo Ham
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Yong Sun Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
32
|
Detecting New Allies: Modifier Screen Identifies a Genetic Interaction Between Imaginal disc growth factor 3 and combover, a Rho-kinase Substrate, During Dorsal Appendage Tube Formation in Drosophila. G3-GENES GENOMES GENETICS 2020; 10:3585-3599. [PMID: 32855169 PMCID: PMC7534437 DOI: 10.1534/g3.120.401476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Biological tube formation underlies organ development and, when disrupted, can cause severe birth defects. To investigate the genetic basis of tubulogenesis, we study the formation of Drosophila melanogaster eggshell structures, called dorsal appendages, which are produced by epithelial tubes. Previously we found that precise levels of Drosophila Chitinase-Like Proteins (CLPs), encoded by the Imaginal disc growth factor (Idgf) gene family, are needed to regulate dorsal-appendage tube closure and tube migration. To identify factors that act in the Idgf pathway, we developed a genetic modifier screen based on the finding that overexpressing Idgf3 causes dorsal appendage defects with ∼50% frequency. Using a library of partially overlapping heterozygous deficiencies, we scanned chromosome 3L and found regions that enhanced or suppressed the Idgf3-overexpression phenotype. Using smaller deletions, RNAi, and mutant alleles, we further mapped five regions and refined the interactions to 58 candidate genes. Importantly, mutant alleles identified combover (cmb), a substrate of Rho-kinase (Rok) and a component of the Planar Cell Polarity (PCP) pathway, as an Idgf3-interacting gene: loss of function enhanced while gain of function suppressed the dorsal appendage defects. Since PCP drives cell intercalation in other systems, we asked if cmb/+ affected cell intercalation in our model, but we found no evidence of its involvement in this step. Instead, we found that loss of cmb dominantly enhanced tube defects associated with Idgf3 overexpression by expanding the apical area of dorsal appendage cells. Apical surface area determines tube volume and shape; in this way, Idgf3 and cmb regulate tube morphology.
Collapse
|
33
|
Decrease in Chitinase 3-Like Protein 1 Levels Reflects Improvement in Liver Fibrosis after HCV Eradication. DISEASE MARKERS 2020; 2020:8539804. [PMID: 33082884 PMCID: PMC7556050 DOI: 10.1155/2020/8539804] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 08/20/2020] [Accepted: 09/05/2020] [Indexed: 12/13/2022]
Abstract
Aim The success of direct-acting antivirals (DAAs) against hepatitis C virus is a major breakthrough in hepatology. Previous studies have shown that chitinase 3-like protein 1 (CHI3L1) was a marker for staging of liver fibrosis caused by HCV. In this investigation, we used CHI3L1 as a surrogate marker to compare dynamic hepatic fibrosis variations following the elimination of HCV among cases receiving sofosbuvir (SOF)-based regimens and pegylated interferon/ribavirin (PR) treatments. Methods The study enrolled 105 patients, including 46 SOF-based regimens treated patients, 34 PR-experienced patients, and 25 untreated patients. Serum samples and clinical data were obtained at the baseline, the end of treatment, and at weeks 24 and 48 after treatments. Results First, we found that serum level of CHI3L1 correlated moderately but significantly with LSM (r = 0.615, P < 0.001) at the baseline, and diagnosed liver cirrhosis at baseline with high accuracy (AUC = 0.939) by ROC analysis. So we explored CHI3L1 as a sensitive biomarker to monitor the regression of liver fibrosis after HCV eradication. We found that the serum CHI3L1 level of CHC cases receiving SOF-based regimen treatments was markedly reduced immediately after treatment compared with that at the baseline (123.79 (118.55) vs. 118.20 (103.68), P = 0.001). For cases undergoing PR treatment, the serum CHI3L1 decreased significantly at week 24 posttreatment compared with that at the baseline (69.98 (51.44) vs 89.15 (110.59), P = 0.016). For the untreated cirrhotic patients, CHI3L1 levels increased at week 96 follow-up compared with that at the baseline (194.73 (172.46) vs. 89.50 (242.97), P = 0.048), reflecting continued worsening of liver fibrosis. Conclusion CHI3L1 is suggested to be the sensitive marker to monitor fibrosis variations in weeks during treatments and after achieving SVR. It has the potential to allow the identification of early treatment failure for a timely switch to alternative treatment and to allow monitoring progression of fibrosis as a risk factor for liver cirrhosis.
Collapse
|
34
|
Selection and Characterization of YKL-40-Targeting Monoclonal Antibodies from Human Synthetic Fab Phage Display Libraries. Int J Mol Sci 2020; 21:ijms21176354. [PMID: 32883029 PMCID: PMC7504393 DOI: 10.3390/ijms21176354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
YKL-40, also known as chitinase-3-like 1 (CHI3L1), is a glycoprotein that is expressed and secreted by various cell types, including cancers and macrophages. Due to its implications for and upregulation in a variety of diseases, including inflammatory conditions, fibrotic disorders, and tumor growth, YKL-40 has been considered as a significant therapeutic biomarker. Here, we used a phage display to develop novel monoclonal antibodies (mAbs) targeting human YKL-40 (hYKL-40). Human synthetic antibody phage display libraries were panned against a recombinant hYKL-40 protein, yielding seven unique Fabs (Antigen-binding fragment), of which two Fabs (H1 and H2) were non-aggregating and thermally stable (75.5 °C and 76.5 °C, respectively) and had high apparent affinities (KD = 2.3 nM and 4.0 nM, respectively). Reformatting the Fabs into IgGs (Immunoglobulin Gs) increased their apparent affinities (notably, for H1 and H2, KD = 0.5 nM and 0.3 nM, respectively), presumably due to the effects of avidity, with little change to their non-aggregation property. The six anti-hYKL-40 IgGs were analyzed using a trans-well migration assay in vitro, revealing that three clones (H1, H2, and H4) were notably effective in reducing cell migration from both A549 and H460 lung cancer cell lines. The three clones were further analyzed in an in vivo animal test that assessed their anti-cancer activities, demonstrating that the tumor area and the number of tumor nodules were significantly reduced in the lung tissues treated with H1 (IgG). Given its high affinity and desirable properties, we expect that the H1 anti-hYKL-40 mAb will be a suitable candidate for developing anti-cancer therapeutics.
Collapse
|
35
|
Saleh AA, Alhanafy AM, Elbahr O, El-Hefnawy SM. Chitinase 3-like 1 gene (T/C) polymorphism and serum YKL-40 levels in patients with hepatocellular carcinoma. Meta Gene 2020; 24:100686. [DOI: https:/doi.org/10.1016/j.mgene.2020.100686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
36
|
Chitinase 3-like 1 gene (T/C) polymorphism and serum YKL-40 levels in patients with hepatocellular carcinoma. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
37
|
Ansari KI, Bhan A, Liu X, Chen MY, Jandial R. Astrocytic IGFBP2 and CHI3L1 in cerebrospinal fluid drive cortical metastasis of HER2+breast cancer. Clin Exp Metastasis 2020; 37:401-412. [PMID: 32279122 DOI: 10.1007/s10585-020-10032-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 04/02/2020] [Indexed: 12/17/2022]
Abstract
The brain is often reported as the first site of recurrence among breast cancer patients overexpressing human epidermal growth factor receptor 2 (HER2). Although most HER2+tumors metastasize to the subcortical region of the brain, a subset develops in the cortical region. We hypothesize that factors in cerebrospinal fluid (CSF) play a critical role in the adaptation, proliferation, and establishment of cortical metastases. We established novel cell lines using patient biopsies to model breast cancer cortical and subcortical metastases. We assessed the localization and growth of these cells in vivo and proliferation and apoptosis in vitro under various conditions. Proteomic analysis of human CSF identified astrocyte-derived factors that support the proliferation of cortical metastases, and we used neutralizing antibodies to test the effects of inhibiting these factors both in vivo and in vitro. The cortical breast cancer brain metastatic cells exhibited greater proliferation than subcortical breast cancer brain metastatic cells in CSF containing several growth factors that nourish both the CNS and tumor cells. Specifically, the astrocytic paracrine factors IGFBP2 and CHI3LI promoted the proliferation of cortical metastatic cells and the formation of metastatic lesions. Disruption of these factors suppressed astrocyte-tumor cell interactions in vitro and the growth of cortical tumors in vivo. Our findings suggest that inhibition of IGFBP2 and CHI3LI signaling, in addition to existing treatment modalities, may be an effective therapeutic strategy targeting breast cancer cortical metastasis.
Collapse
Affiliation(s)
- Khairul I Ansari
- Division of Neurosurgery, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
- Celcuity, 16305 36th Ave N, Suite 100, Minneapolis, MN, 55446, USA.
| | - Arunoday Bhan
- Division of Neurosurgery, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Xueli Liu
- Division of Biostatistics, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Mike Y Chen
- Division of Neurosurgery, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Rahul Jandial
- Division of Neurosurgery, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
38
|
Mhaidly R, Mechta-Grigoriou F. Fibroblast heterogeneity in tumor micro-environment: Role in immunosuppression and new therapies. Semin Immunol 2020; 48:101417. [PMID: 33077325 DOI: 10.1016/j.smim.2020.101417] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023]
|
39
|
Park KR, Yun HM, Yoo K, Ham YW, Han SB, Hong JT. Chitinase 3 like 1 suppresses the stability and activity of p53 to promote lung tumorigenesis. Cell Commun Signal 2020; 18:5. [PMID: 32127023 PMCID: PMC7055043 DOI: 10.1186/s12964-019-0503-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/26/2019] [Indexed: 12/22/2022] Open
Abstract
Background Chitinase 3 like 1 protein (Chi3L1) is expressed in several cancers, and a few evidences suggest that the secreted Chi3L1 contributes to tumor development. However, the molecular mechanisms of intracellular Chi3L1 are unknown in the lung tumor development. Methods: In the present study, we generated Chi3L1 knockout mice (Chi3L1KO(−/−)) using CRISPR/Cas9 system to investigate the role of Chi3L1 on lung tumorigenesis. Results We established lung metastasis induced by i.v. injections of B16F10 in Chi3L1KO(−/−). The lung tumor nodules were significantly reduced in Chi3L1KO(−/−) and protein levels of p53, p21, BAX, and cleaved-caspase 3 were significantly increased in Chi3L1KO(−/−), while protein levels of cyclin E1, CDK2, and phsphorylation of STAT3 were decreased in Chi3L1KO(−/−). Allograft mice inoculated with B16F10 also suppressed tumor growth and increased p53 and its target proteins including p21 and BAX. In addition, knockdown of Chi3L1 in lung cancer cells inhibited lung cancer cell growth and upregulated p53 expression with p21 and BAX, and a decrease in phosphorylation of STAT3. Furthermore, we found that intracellular Chi3L1 physically interacted and colocalized with p53 to inhibit its protein stability and transcriptional activity for target genes related with cell cycle arrest and apoptosis. In lung tumor patient, we clinically found that Chi3L1 expression was upregulated with a decrease in p53 expression, as well as we validated that intracellular Chi3L1 was colocalized, reversely expressed, and physically interacted with p53, which results in suppression of the expression and function of p53 in lung tumor patient. Conclusions Our studies suggest that intracellular Chi3L1 plays a critical role in the lung tumorigenesis by regulating its novel target protein, p53 in both an in vitro and in vivo system.
Collapse
Affiliation(s)
- Kyung-Ran Park
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul, 02453, Republic of Korea
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul, 02453, Republic of Korea
| | - Kyeongwon Yoo
- KRIBB/Bio-venture Center 113 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Young Wan Ham
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Sang Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| |
Collapse
|
40
|
Jiang Z, Wang S, Jin J, Ying S, Chen Z, Zhu D, Xiao B, Hu Y, Qian Y, Cai T, Fu L. The clinical significance of serum chitinase 3-like 1 in hepatitis B-related chronic liver diseases. J Clin Lab Anal 2020; 34:e23200. [PMID: 31916309 PMCID: PMC7246367 DOI: 10.1002/jcla.23200] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/01/2019] [Accepted: 12/14/2019] [Indexed: 02/07/2023] Open
Abstract
Aim In the present study, we purposed to determine serum chitinase 3‐like 1 (CHI3L1) expression characteristics in chronic liver diseases monoinfected with hepatitis B virus and analyze its diagnostic value in liver fibrosis. Methods A total of 467 chronic hepatitis B (CHB) patients, 312 liver cirrhosis (LC) patients, and 104 hepatocellular carcinoma (HCC) patients at our institution were enrolled, and clinical indicators were analyzed. Results Our data have shown that the expression level of serum CHI3L1 was steadily increased from CHB to LC to HCC (P < .001). Serum CHI3L1 expression levels were positively associated with liver stiffness measurement (LSM), fibrosis‐4 (FIB‐4) index, aspartate aminotransferase‐to‐platelet ratio index (APRI), and HCC stage. The receiver operating characteristic (ROC) curve proved that serum CHI3L1 was superior to other noninvasive methods (LSM, FIB‐4, and APRI) with an area under the ROC curve (AUC) of 0.97 in diagnosing significant fibrosis. Conclusions Serum CHI3L1 harbors significant clinical value in chronic liver diseases infected with hepatitis B virus, especially in the diagnosis of fibrosis.
Collapse
Affiliation(s)
- Zhenluo Jiang
- Department of Hepatology, HwaMei Hospital, University Of Chinese Academy Of Sciences, Ningbo, China.,Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| | - Shuwei Wang
- Department of Hepatology, HwaMei Hospital, University Of Chinese Academy Of Sciences, Ningbo, China.,Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| | - Jiancheng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| | - Sheng Ying
- Department of Hepatology, HwaMei Hospital, University Of Chinese Academy Of Sciences, Ningbo, China
| | - Zhigang Chen
- Department of Hepatology, HwaMei Hospital, University Of Chinese Academy Of Sciences, Ningbo, China
| | - Dedong Zhu
- Department of Hepatology, HwaMei Hospital, University Of Chinese Academy Of Sciences, Ningbo, China
| | - Bingxiu Xiao
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| | - Yaoren Hu
- Department of Hepatology, HwaMei Hospital, University Of Chinese Academy Of Sciences, Ningbo, China.,Key Laboratory of Diagnosis and Treatment of Digestive System tumors of Zhejiang Province, Ningbo, China
| | - Yunsong Qian
- Department of Hepatology, HwaMei Hospital, University Of Chinese Academy Of Sciences, Ningbo, China
| | - Ting Cai
- Key Laboratory of Diagnosis and Treatment of Digestive System tumors of Zhejiang Province, Ningbo, China
| | - Liyun Fu
- Department of Hepatology, HwaMei Hospital, University Of Chinese Academy Of Sciences, Ningbo, China.,Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China.,Key Laboratory of Diagnosis and Treatment of Digestive System tumors of Zhejiang Province, Ningbo, China
| |
Collapse
|
41
|
Park KR, Yun HM, Hong JT. G721-0282 inhibits cell growth and induces apoptosis in human osteosarcoma through down-regulation of the STAT3 pathway. Int J Biol Sci 2020; 16:330-341. [PMID: 31929760 PMCID: PMC6949149 DOI: 10.7150/ijbs.37781] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma (OS) is considered the most common type of primary malignant bone tumor, which has an urgent need for more effective treatment. Recently, chitinase 3 like 1 (Chi3L1) expression has been found in a variety of cancer cells. However it is not known whether Chi3L1 regulates the STAT3 pathway in OS cells. Herein, we examined the effects of the G721-0282, a ligand of Chi3L1, in vitro and in vivo against OS cells. G721-0282 inhibited the proliferation of OS cells and induced apoptosis. This apoptosis was accompanied by upregulation of apoptotic proteins (PARP and procaspase-3), but downregulation of anti-apoptotic proteins (Survivin and Bcl-2). G721-0282 induced the inactivation of mitogen-activated protein kinases (MAPKs) with a decrease in the phosphorylation of Src and STAT3 in OS cells. Importantly, overexpression of Chi3L1 potentiated the effects of G721-0282, while knockdown of Chi3L1 attenuated the effects of G721-0282. Docking model study also showed that G721-0282 interacted with Chi3L1. In addition, G721-0282 inhibited cell migration, invasion, and colony formation. Furthermore, the anti-tumor effects of G721-0282 were observed in an xenograft in vivo model in association with the reduced expression of Chi3L1, PCNA, Cyclin D1, p-STAT3, as well as the increased expression of Chi3L1 was correlated with the p-STAT3 level in human bone tumor tissues. Taken together, a Chi3L1 ligand, G721-0282 may be an attractive therapeutic strategy for OS, especially in vitro and in vivo anti-proliferative effects against OS cells through the inhibition of the STAT3 pathway, and suggest the potentially therapeutic application of G721-0282 in the treatment of OS.
Collapse
Affiliation(s)
- Kyung-Ran Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 194-31, Republic of Korea
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 194-31, Republic of Korea
| |
Collapse
|
42
|
Kim EG, Kim MN, Hong JY, Lee JW, Kim SY, Kim KW, Lee CG, Elias JA, Song TW, Sohn MH. Chitinase 3-Like 1 Contributes to Food Allergy via M2 Macrophage Polarization. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 12:1012-1028. [PMID: 32935492 PMCID: PMC7492506 DOI: 10.4168/aair.2020.12.6.1012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022]
Abstract
Purpose Food allergy is a hypersensitive immune response to specific food proteins. Chitinase 3-like 1 (CHI3L1, also known as YKL-40 in humans or BRP-39 in mice) is associated with various chronic diseases, such as cancer, rheumatoid arthritis, and allergic disease. CHI3L1 is involved in allergen sensitization and type 2 helper T (Th2) inflammation, but the role of CHI3L1 in food allergy remains unclear. In this study, we sought to investigate the role of CHI3L1 in the development of food allergy. Methods We measured serum levels of CHI3L1 in food allergic patients. Food allergy was induced in wild-type (WT) and CHI3L1 null mutant (CHI3L1−/−) BALB/c mice with ovalbumin (OVA). We investigated Th2 immune responses, M2 macrophage polarization, and mitogen-activated protein kinase (MAPK)/phosphoinositide 3-kinase (PI3K) signaling pathways, and also performed transcriptome analysis. Results Serum levels of CHI3L1 were significantly higher in children with food allergy compared with those in healthy controls. Furthermore, CHI3L1 expression levels were elevated in WT mice after OVA treatment. Food allergy symptoms, immunoglobulin E levels, Th2 cytokine production, and histological injury were attenuated in food allergy-induced CHI3L1−/− mice compared with those in food allergy-induced WT mice. CHI3L1 expression was increased in OVA-treated WT intestinal macrophages and caused M2 macrophage polarization. Furthermore, CHI3L1 was involved in the extracellular signal-regulated kinases (ERK) and AKT signaling pathways and was associated with immune response and lipid metabolism as determined through transcriptome analysis. Conclusions CHI3L1 plays a pivotal role in Th2 inflammation and M2 macrophage polarization through MAPK/ERK and PI3K/AKT phosphorylation in food allergy.
Collapse
Affiliation(s)
- Eun Gyul Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Na Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Yeon Hong
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Woo Lee
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Yeon Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Won Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA.,Department of Internal Medicine, Hanyang University, Seoul, Korea
| | - Jack A Elias
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Tae Won Song
- Department of Pediatrics, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea.
| | - Myung Hyun Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
43
|
Hübner K, Karwelat D, Pietsch E, Beinborn I, Winterberg S, Bedenbender K, Benedikter BJ, Schmeck B, Vollmeister E. NF-κB-mediated inhibition of microRNA-149-5p regulates Chitinase-3-like 1 expression in human airway epithelial cells. Cell Signal 2019; 67:109498. [PMID: 31837465 DOI: 10.1016/j.cellsig.2019.109498] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/22/2022]
Abstract
Lower respiratory tract infections are among the most common causes of death worldwide. Main pathogens leading to these severe infections are viruses and gram-positive bacteria that activate toll-like receptor (TLR)-mediated immune responses via pathogen-associated molecular patterns. One protective factor induced during infection is Chitinase-3-like 1 (CHI3L1), which exerts various functions, e.g. in host cell proliferation and bacterial counteraction, and has been proposed as a biomarker in several acute and chronic inflammatory conditions. MicroRNAs (miR) have become important regulators of inflammation and infection and are considered therapeutic targets in recent years. However, it is not known whether microRNAs play a role in the regulation of CHI3L1 expression in TLR-mediated respiratory epithelial cell inflammation. In this study, we analysed the pre- and post-transcriptional regulation of CHI3L1 by TLRs in bronchial epithelial cells. Therefore, we stimulated BEAS-2B cells with the bacterial TLR2-ligand lipoteichoic acid or the viral dsRNA analogue poly(I:C). We observed an increase in the expression of CHI3L1, which was dependent on TNF-α-mediated NF-κB activation in TLR2- and TLR3-activated cells. Moreover, TLR2 and - 3 stimulation caused downregulation of the microRNA miR-149-5p, an effect that could be suppressed by inhibiting NF-κB translocation into the nucleus. Luciferase reporter assays identified a direct interaction of miR-149-5p with the CHI3L1 3´untranslated region. This interaction was confirmed by inhibition and overexpression of miR-149-5p in BEAS-2B cells, which altered the expression levels of CHI3L1 mRNA. In summary, miR-149-5p directly regulates CHI3L1 in context of TLR-mediated airway epithelial cell inflammation and may be a potential therapeutic target in inflammation and other diseases.
Collapse
Affiliation(s)
- Kathleen Hübner
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany
| | - Diana Karwelat
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany
| | - Emma Pietsch
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany
| | - Isabell Beinborn
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany
| | - Sarah Winterberg
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany
| | - Katrin Bedenbender
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany
| | - Birke J Benedikter
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany; Department of Medical Microbiology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, PO box 5800, 6202AZ Maastricht, the Netherlands
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany; Department of Pulmonary and Critical Care Medicine, University Medical Center Marburg, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany.
| | - Evelyn Vollmeister
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Hans-Meerwein Straße 2, 35043 Marburg, Hesse, Germany.
| |
Collapse
|
44
|
Yin S, Wang N, Riabov V, Mossel DM, Larionova I, Schledzewski K, Trofimova O, Sevastyanova T, Zajakina A, Schmuttermaier C, Gratchev A, Flatley A, Kremmer E, Zavyalova M, Cherdyntseva N, Simon-Keller K, Marx A, Klüter H, Goerdt S, Kzhyshkowska J. SI-CLP inhibits the growth of mouse mammary adenocarcinoma by preventing recruitment of tumor-associated macrophages. Int J Cancer 2019; 146:1396-1408. [PMID: 31525266 DOI: 10.1002/ijc.32685] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 06/30/2019] [Accepted: 08/07/2019] [Indexed: 02/05/2023]
Abstract
Chitinase-like proteins (CLP) are chitin-binding proteins that lack chitin hydrolyzing activity, but possess cytokine-like and growth factor-like properties, and play crucial role in intercellular crosstalk. Both human and mice express two members of CLP family: YKL-40 and stabilin-1 interacting chitinase-like protein (SI-CLP). Despite numerous reports indicating the role of YKL-40 in the support of angiogenesis, tumor cell proliferation, invasion and metastasis, the role of its structurally related protein SI-CLP in cancer was not reported. Using gain-of-function approach, we demonstrate in the current study that the expression of recombinant SI-CLP in mouse TS/A mammary adenocarcinoma cells results in significant and persistent inhibition of in vivo tumor growth. Using quantitative immunohistochemistry, we show that on the cellular level this phenomenon is associated with reduced infiltration of tumor-associated macrophages (TAMs), CD4+ and FoxP3+ cells in SI-CLP expressing tumors. Gene expression analysis in TAM isolated from SI-CLP-expressing and control tumors demonstrated that SI-CLP does not affect macrophage phenotype. However, SI-CLP significantly inhibited migration of murine bone-marrow derived macrophages and human primary monocytes toward monocyte-recruiting chemokine CCL2 produced in the tumor microenvironment (TME). Mechanistically, SI-CLP did not affect CCL2/CCR2 interaction, but suppressed cytoskeletal rearrangements in response to CCL2. Altogether, our data indicate that SI-CLP functions as a tumor growth inhibitor in mouse breast cancer by altering cellular composition of TME and blocking cytokine-induced TAM recruitment. Taking into consideration weak to absent expression of SI-CLP in human breast cancer, it can be considered as a therapeutic protein to block TAM-mediated support of breast tumor growth.
Collapse
Affiliation(s)
- Shuiping Yin
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Nan Wang
- Department of Dermatology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science andTechnology, Wuhan, China
| | - Vladimir Riabov
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| | - Dieuwertje M Mossel
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia.,Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Kai Schledzewski
- Department of Dermatology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Olga Trofimova
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Tatyana Sevastyanova
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Anna Zajakina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Christina Schmuttermaier
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Alexei Gratchev
- Department of Dermatology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Laboratory for Tumor Stromal Cell Biology, Institute of Carcinogenesis, NN Blokhin Cancer Research Center, Russian Academy of Sciences, Moscow, Russia
| | - Andrew Flatley
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Marina Zavyalova
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Nadezhda Cherdyntseva
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia.,Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Katja Simon-Keller
- Institute of Pathology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Alexander Marx
- Institute of Pathology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Harald Klüter
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Sergij Goerdt
- Department of Dermatology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Julia Kzhyshkowska
- Medical Faculty Mannheim, Institute for Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia.,German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| |
Collapse
|
45
|
Zhou YJ, Ye FZ, Li YY, Pan XY, Chen YX, Wu XX, Xiong JJ, Liu WY, Xu SH, Chen YP, Zheng MH. Individualized risk prediction of significant fibrosis in non-alcoholic fatty liver disease using a novel nomogram. United European Gastroenterol J 2019; 7:1124-1134. [PMID: 31662869 PMCID: PMC6794687 DOI: 10.1177/2050640619868352] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/08/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Fibrosis is deemed to be a pivotal determinant of the long-term prognosis in non-alcoholic fatty liver disease (NAFLD). OBJECTIVE We aimed to develop a novel nomogram-based non-invasive model to accurately predict significant fibrosis in patients with NAFLD. METHODS We designed a prospective cohort study including 207 patients with biopsy-proven NAFLD. Detailed anthropometric and fibrosis-related laboratory parameters were collected. A nomogram was established based on variables that were independently associated with significant fibrosis identified by the logistic regression model. Then it was compared with aspartate aminotransferase-to-platelet ratio index (APRI), NAFLD fibrosis score (NFS), FIB-4 and BARD score. Diagnostic accuracy was assessed according to area under the receiver operator characteristic curve (AUROC), sensitivity, specificity, positive and negative predictive values, and decision curve analysis. RESULTS Variables included in the nomogram were: waist-to-height ratio, hyaluronic acid, procollagen-III-peptide, chitinase-3-like protein 1, and cytokeratine-18 neoepitope M65. The discrimination ability of the nomogram (AUROC = 0.829, 95%CI 0.755-0.904) was significantly superior to APRI (AUROC = 0.670, 95%CI 0.563-0.777), NFS (AUROC = 0.601, 95%CI 0.480-0.722), FIB-4 (AUROC = 0.624, 95%CI 0.511-0.736) and BARD (AUROC = 0.579, 95%CI 0.459-0.699) for significant fibrosis (all p < 0.05). The nomogram showed a larger net benefit to aid in decision-making as to whether biopsy is required. CONCLUSIONS This novel nomogram was more accurate, and achieved higher net benefit than APRI, NFS, FIB-4 and BARD to detect significant fibrosis. It can be useful as a non-invasive method to screen ≥F2 fibrosis in the overall population with NAFLD.
Collapse
Affiliation(s)
- Yu-Jie Zhou
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Fang-Zhou Ye
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yang-Yang Li
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Yan Pan
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying-Xiao Chen
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xi-Xi Wu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiao-Jiao Xiong
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wen-Yue Liu
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shi-Hao Xu
- Department of Ultrasonography, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yong-Ping Chen
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
46
|
Sex difference in CHI3L1 expression levels in human brain aging and in Alzheimer’s disease. Brain Res 2019; 1720:146305. [DOI: 10.1016/j.brainres.2019.146305] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/20/2019] [Accepted: 06/23/2019] [Indexed: 02/07/2023]
|
47
|
Astrogliosis Releases Pro-Oncogenic Chitinase 3-Like 1 Causing MAPK Signaling in Glioblastoma. Cancers (Basel) 2019; 11:cancers11101437. [PMID: 31561550 PMCID: PMC6826948 DOI: 10.3390/cancers11101437] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/21/2019] [Accepted: 09/24/2019] [Indexed: 12/15/2022] Open
Abstract
Although reactive astrocytes constitute a major component of the cellular environment in glioblastoma, their function and crosstalk to other components of the environment is still poorly understood. Gene expression analysis of purified astrocytes from both the tumor core and non-infiltrated cortex reveals a tumor-related up-regulation of Chitinase 3-like 1 (CHI3L1), a cytokine which is related to inflammation, extracellular tissue remodeling, and fibrosis. Further, we established and validated a co-culture model to investigate the impact of reactive astrocytes within the tumor microenvironment. Here we show that reactive astrocytes promote a subtype-shift of glioblastoma towards the mesenchymal phenotype, driving mitogen-activated protein kinases (MAPK) signaling as well as increased proliferation and migration. In addition, we demonstrate that MAPK signaling is directly caused by a CHI3L1-IL13RA2 co-binding, which leads to increased downstream MAPK and AKT signaling. This novel microenvironmental crosstalk highlights the crucial role of non-neoplastic cells in malignant brain tumors and opens up new perspectives for targeted therapies in glioblastoma.
Collapse
|
48
|
Li F, Sheng Z, Lan H, Xu J, Li J. Downregulated CHI3L1 alleviates skeletal muscle stem cell injury in a mouse model of sepsis. IUBMB Life 2019; 72:214-225. [PMID: 31463997 DOI: 10.1002/iub.2156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 08/05/2019] [Indexed: 01/03/2023]
Abstract
Sepsis is an acute systemic inflammatory response of the body to microbial infection and a life-threatening condition associated with multiple organ failure. Recent data suggest that sepsis survivors present with long-term myopathy due to the dysfunction of skeletal muscle stem cells and satellite cells. Accumulating studies have implicated chitinase-3-like-1 protein (CHI3L1) in a variety of infectious diseases, specifically sepsis. Therefore, the aim of the present study is to elucidate the potential mechanism by which CHI3L1 is involved in the injury of skeletal muscle stem cells in mouse models of sepsis. An in vitro cell model was developed by lipopolysaccharide (LPS) and in vivo mouse model of sepsis was induced by CRP-like protein (CLP). To elucidate the biological significance behind the silencing of CHI3L1, modeled skeletal muscle stem cells and mice were treated with siRNA against CHI3L1 or overexpressed CHI3L1. Highly expressed CHI3L1 was found in skeletal muscle tissues of mice with sepsis. Besides, siRNA-mediated silencing of CHI3L1 was revealed to increase Bcl-2 expression along with cell proliferation, while diminishing Bax expression, cell apopstosis as well as serum levels of TNF-α, IL-1β, INF-γ, IL-10, and IL-6. Taken conjointly, this present study provided evidence suggesting that downregulation of CHI3L1 has the potential to prevent the injury of skeletal muscle stem cells in mice with sepsis. Collectively, CHI3L1 may serve as a valuable therapeutic strategy in alleviating sepsis.
Collapse
Affiliation(s)
- Fuxing Li
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Zhiyong Sheng
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Haibing Lan
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jianning Xu
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
49
|
Yeo IJ, Lee CK, Han SB, Yun J, Hong JT. Roles of chitinase 3-like 1 in the development of cancer, neurodegenerative diseases, and inflammatory diseases. Pharmacol Ther 2019; 203:107394. [PMID: 31356910 DOI: 10.1016/j.pharmthera.2019.107394] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2019] [Indexed: 02/07/2023]
Abstract
Chitinase 3-like 1 (CHI3L1) is a secreted glycoprotein that mediates inflammation, macrophage polarization, apoptosis, and carcinogenesis. The expression of CHI3L1 is strongly increased by various inflammatory and immunological conditions, including rheumatoid arthritis, multiple sclerosis, Alzheimer's disease, and several cancers. However, its physiological and pathophysiological roles in the development of cancer and neurodegenerative and inflammatory diseases remain unclear. Several studies have reported that CHI3L1 promotes cancer proliferation, inflammatory cytokine production, and microglial activation, and that multiple receptors, such as advanced glycation end product, syndecan-1/αVβ3, and IL-13Rα2, are involved. In addition, the pro-inflammatory action of CHI3L1 may be mediated via the protein kinase B and phosphoinositide-3 signaling pathways and responses to various pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, interleukin-6, and interferon-γ. Therefore, CHI3L1 could contribute to a vast array of inflammatory diseases. In this article, we review recent findings regarding the roles of CHI3L1 and suggest therapeutic approaches targeting CHI3L1 in the development of cancers, neurodegenerative diseases, and inflammatory diseases.
Collapse
Affiliation(s)
- In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Chong-Kil Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
50
|
Zadi Heydarabad M, Baharaghdam S, Azimi A, Mohammadi H, Eivazi Ziaei J, Yazdanpanah B, Zak MS, Farahani ME, Dohrabpour A, Partash N, Talebi M. The role of tumor suppressor of resveratrol and prednisolone by downregulation of YKL-40 expression in CCRF-CEM cell line. J Cell Biochem 2018; 120:3773-3779. [PMID: 30426549 DOI: 10.1002/jcb.27659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/21/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND Acute lymphoblastic leukemia (ALL) is characterized by excessive accumulation of lymphoblast and progenitors. Leukemia is the most common cancer in children and ALL is the most common subtype. Many studies have shown that the YKL-40 gene is one of the most widely expressed genes in tumors, including leukemia, but not in healthy blood cells. Clinical studies have shown that serum YKL-40 levels have a positive correlation with tumor expansion, in addition to being a prognostic agent independent of a short relapse-free interval, as well as a brief overall survival in patients with various cancers. The previous study shows that YKL-40 is closely related to the degree of pathology or degree of human leukemia pathology and plays an important role in cell proliferation. Hence, the YKL-40 can be an attractive target in designing anticancer therapies. METHODS CCRF-CEM cells were treated with resveratrol and prednisolone. For analysis of YKL-40 expression changes under medication, real-time polymerase chain reaction (PCR) and Western blot techniques were used at resonating intervals of 24 and 48 hours. RESULTS The effect of 15, 50, and 100 μM resveratrol and 700 μM of prednisolone on CCRF-CEM cells reduced YKL-40. The YKL-40 gene was quantitatively measured using RT-PCR. The Western blot method was used to evaluate changes in the expression of YKL-40 protein. CONCLUSION In this study, we first evaluated YKL-40 expression and resveratrol and prednisolone effect on YKL-40 in ALL. This finding supports the idea of targeting YKL-40 as a new drug treatment of ALL and extends the use of resveratrol in antileukemia research.
Collapse
Affiliation(s)
| | - Sina Baharaghdam
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ako Azimi
- Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Hamed Mohammadi
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jamal Eivazi Ziaei
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behruz Yazdanpanah
- Department of Laboratory Sciences, School Paramedics, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohsen Sharif Zak
- Clinical Biochemistry and Laboratory, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Aghdas Dohrabpour
- Department of Microbiology, Yasuj Branch, Islamic Azad University, Tehran, Iran
| | - Nasim Partash
- Department of Nursing, School of Nursing and Midwifery, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mehdi Talebi
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.,Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|