1
|
Dora D, Revisnyei P, Pasic A, Galffy G, Dulka E, Mihucz A, Roskó B, Szincsak S, Iliuk A, Weiss GJ, Lohinai Z. Host and bacterial urine proteomics might predict treatment outcomes for immunotherapy in advanced non-small cell lung cancer patients. Front Immunol 2025; 16:1543817. [PMID: 40297587 PMCID: PMC12035445 DOI: 10.3389/fimmu.2025.1543817] [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: 12/11/2024] [Accepted: 03/05/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Urine samples are non-invasive approaches to study potential circulating biomarkers from the host organism. Specific proteins cross the bloodstream through the intestinal barrier and may also derive from gut microbiota. In this study, we aimed to evaluate the predictive role of the host and bacterial urine extracellular vesicle (EV) proteomes in patients with non-small cell lung cancer (NSCLC) treated with anti-PD1 immunotherapy. Methods We analyzed the urine EV proteome of 33 advanced-stage NSCLC patients treated with anti-PD1 immunotherapy with LC-MS/MS, stratifying patients according to long (>6 months) and short (≤6 months) progression-free survival (PFS). Gut microbial communities on a subcohort of 23 patients were also analyzed with shotgun metagenomics. Internal validation was performed using the Random Forest (RF) machine learning (ML) algorithm. RF was validated with a non-linear Bayesian ML model. Gene enrichment, and pathway analysis of host urine proteins were analyzed using the Reactome and Gene Ontology databases. Results We identified human (n=3513), bacterial (n=2647), fungal (n=19), and viral (n=4) proteins. 186 human proteins showed differential abundance (p<0.05) according to PFS groups, 101 being significantly more abundant in patients with short PFS and n=85 in patients with long PFS. We found several pathways that were significantly enriched in patients with short PFS (vs long PFS). Multivariate Cox regression showed that human urine proteins MPP5, IGKV6-21, NT5E, and KRT27 were strongly associated with long PFS, and LMAN2, NUTF2, NID1, TNC, IGF1, BCR, GPHN, and PPBP showed the strongest association with short PFS. We revealed that an increased bacterial/host protein ratio in the urine is more frequent in patients with long PFS. Increased abundance of E. coli and E. faecalis proteins in the urine positively correlates with their gut metagenomic abundance. RF ML model supported the reliability in predicting PFS for critical human urine proteins (AUC=0.89), accuracy (95%) and Bacterial proteins (AUC=0.74). Conclusion To our knowledge, this is the first study to depict the predictive role of the host and bacterial urine proteome in anti-PD1-treated advanced NSCLC.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Peter Revisnyei
- Department of Telecommunications and Media Informatics, Budapest University of Technology and Economics, Budapest, Hungary
- HUN-REN-BME Information Systems Research Group, Budapest, Hungary
| | - Alija Pasic
- Department of Telecommunications and Media Informatics, Budapest University of Technology and Economics, Budapest, Hungary
| | | | - Edit Dulka
- County Hospital of Torokbalint, Torokbalint, Hungary
| | - Anna Mihucz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Brigitta Roskó
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Sara Szincsak
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, IN, United States
| | - Glen J. Weiss
- Department of Medicine, UMass Chan Medical School, Worcester, MA, United States
| | - Zoltan Lohinai
- County Hospital of Torokbalint, Torokbalint, Hungary
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| |
Collapse
|
2
|
Lee JH, Son S, Ko Y, Lim H, Lee M, Kang MG, Kim H, Lee KM, Shin I. Nidogen-1 suppresses cell proliferation, migration, and glycolysis via integrin β1-mediated HIF-1α downregulation in triple-negative breast cancer. Sci Rep 2025; 15:10633. [PMID: 40148359 PMCID: PMC11950294 DOI: 10.1038/s41598-024-84880-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 12/27/2024] [Indexed: 03/29/2025] Open
Abstract
Nidogen-1 (NID1) is a secreted glycoprotein widely distributed in basement membranes. NID1 interacts with extracellular matrix proteins such as collagen and laminin and has been implicated in the progression of various cancers. However, study on the role of NID1 in breast cancer is scarce and inconsistent. In this work, we found that the expression of NID1 is significantly lower in breast cancer tissue than in normal tissue. In addition, NID1 expression correlated negatively with a poor prognosis for breast cancer patients. Based on those findings, we speculated that NID1 might act as a cancer suppressor in breast cancer. To investigate the role of NID1 in breast cancer, we constructed NID1-overexpressing cell lines. NID1 overexpression decreased breast cancer cell proliferation, migration, and in vivo tumor growth. Moreover, glucose metabolism, which is known to enhance cancer cell proliferation and migration, was also decreased by NID1 overexpression. Mechanistically, NID1 overexpression downregulated hypoxia-inducible factor-1α (HIF-1α) expression at the transcription level. Furthermore, we found that NID1 reduced integrin β1 stability and downregulated the transcription of HIF-1α through the FAK/Src/NF-κB p65 signaling axis, which is downstream of integrin β1. Together, the results of this study demonstrate the tumor suppressive role of NID1 in triple-negative breast cancer.
Collapse
Affiliation(s)
- Joo-Hyung Lee
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Seogho Son
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Yunhyo Ko
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Hogeun Lim
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Minhyeok Lee
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Min-Gyeong Kang
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Hyungjoo Kim
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Kyung-Min Lee
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Incheol Shin
- Department of Life Science, Hanyang University, Seoul, 04763, Korea.
- Natural Science Institute, Hanyang University, Seoul, 04763, Korea.
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, 04763, Korea.
| |
Collapse
|
3
|
Xue T, Yeung CLS, Mao X, Tey SK, Lo KW, Tang AHN, Yun JP, Yam JWP. Development of a broadly potent neutralizing antibody targeting Nidogen 1 effectively inhibits cancer growth and metastasis in preclinical tumor models. J Transl Int Med 2025; 13:78-92. [PMID: 40115036 PMCID: PMC11921815 DOI: 10.1515/jtim-2025-0008] [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] [Indexed: 03/22/2025] Open
Abstract
Background and Objectives Nidogen 1 (NID1) is a highly conserved structural component of the extracellular matrix (ECM), which interacts with different basement membrane (BM) proteins to form a stabilized meshwork. The promoting ability of NID1 in cancer development and metastasis has been demonstrated in multiple cancer types, including ovarian cancer, breast cancer, and hepatocellular carcinoma (HCC). This suggests that NID1 holds great potential as a therapeutic target for cancer treatment. However, currently, there is a lack of commercially available neutralizing antibody for clinical testing and treatment. Methods To address this, we utilized hybridoma technology to develop a monoclonal neutralizing antibody which targets the critical G2 region of NID1. The therapeutic effect of this NID1 neutralizing antibody against a wide range of human cancer cells was evaluated. Results The results showed that NID1 neutralizing antibody effectively attenuated the growth, motility and metastasis of HCC, lung cancer, breast cancer and nasopharyngeal carcinoma cells in vitro. The proof-of-concept of targeting NID1 using neutralizing antibody was further demonstrated in various animal models. Mechanistically, our findings indicate that treatment with NID1 neutralizing antibody leads to the deregulation of hypoxia-inducible factor-1 (HIF-1α) pathway in cancer cells. Conclusions Taken together, this study offers promising prospects for a new pan-cancer monoclonal antibody-based strategy by targeting the tumor-associated membrane protein NID1.
Collapse
Affiliation(s)
- Tingmao Xue
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Cherlie Lot Sum Yeung
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Sze Keong Tey
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alexander Hin Ning Tang
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jing Ping Yun
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong SAR, China
- DiagnoVEX Therapeutics Limited, Hong Kong SAR, China
| |
Collapse
|
4
|
Li Y, Zhang L, Xu G, Xu G, Chen J, Zhao K, Li M, Jin J, Peng C, Wang K, Pan S, Zhu K. Exploration and validation of a novel reactive oxygen species-related signature for predicting the prognosis and chemotherapy response of patients with bladder cancer. Front Immunol 2024; 15:1493528. [PMID: 39749345 PMCID: PMC11693660 DOI: 10.3389/fimmu.2024.1493528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Background Reactive Oxygen Species (ROS), a hallmark of cancer, is related to prognosis, tumor progression, and treatment response. Nevertheless, the correlation of ROS-based molecular signature with clinical outcome and immune cell infiltration has not been thoroughly studied in bladder cancer (BLCA). Accordingly, we aimed to thoroughly examine the role and prognostic value of ROS-related genes in BLCA. Methods We obtained RNA sequencing and clinical data from The Cancer Genome Atlas (TCGA) for bladder cancer (BLCA) patients and identified ROS-associated genes using the GeneCards and Molecular Signatures Database (MSigDB). We then analyzed differential gene expression between BLCA and normal tissues and explored the functions of these ROS-related genes through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Protein-Protein Interaction (PPI) analysis. Prognostic ROS-related genes were identified using Univariate Cox regression (UCR) and LASSO analyses, which were further refined in a Multivariate Cox Regression (MCR) analysis to develop a Prognostic Signature (PS). This PS was validated in the GSE13507 cohort, assessing its predictive power with Kaplan-Meier survival and time-dependent ROC curves. To forecast BLCA outcomes, we constructed a nomogram integrating the PS with clinical variables. We also investigated the signature's molecular characteristics through Gene Set Enrichment Analysis (GSEA), Immune Cell Infiltration (ICI), and Tumor Mutational Burden (TMB) analyses. The Genomics of Drug Sensitivity in Cancer (GDSC) database was used to predict chemotherapy responses based on the PS. Additionally, we screened for Small-Molecule Drugs (SMDs) targeting ROS-related genes using the CMAP database. Finally, we validated our findings by checking protein levels of the signature genes in the Human Protein Atlas (HPA) and confirmed the role of Aldo-keto reductase family 1 member B1 (AKR1B1) through in vitro experiments. Results The constructed and validated PS that comprised 17 ROS-related genes exhibited good performance in predicting overall survival (OS), constituting an independent prognostic biomarker in BLCA patients. Additionally, we successfully established a nomogram with superior predictive capacity, as indicated by the calibration plots. The bioinformatics analysis findings showcased the implication of PS in several oncogenic pathways besides tumor ICI regulation. The PS was negatively associated with the TMB. The high-risk group patients had greater chemotherapy sensitivity in comparison to low-risk group patients. Further, 11 candidate SMDs were identified for treating BLCA. The majority of gene expression exhibited a correlation with the protein expression. In addition, the expression of most genes was consistent with protein expression. Furthermore, to test the gene reliability we constructed, AKR1B1, one of the seventeen genes identified, was used for in-depth validation. In vitro experiments indicate that siRNA-mediated AKR1B1 silencing impeded BLCA cell viability, migration, and proliferation. Conclusions We identified a PS based on 17 ROS-related genes that represented independent OS prognostic factors and 11 candidate SMDs for BLCA treatment, which may contribute to the development of effective individualized therapies for BLCA.
Collapse
Affiliation(s)
- Yulei Li
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Lulu Zhang
- Medical Research Center, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Gang Xu
- Department of Urology, Nanchang People’s Hospital, Nanchang, China
| | - Gang Xu
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Jiajun Chen
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Keyuan Zhao
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Mengyao Li
- Department of Pathology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Jing Jin
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Chao Peng
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Kaifang Wang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, Macao SAR, China
| | - Shouhua Pan
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Ke Zhu
- Department of Urology, Nanchang People’s Hospital, Nanchang, China
| |
Collapse
|
5
|
Dennis CD, Dillon JT, Patel PH, Cohen DJ, Halquist MS, Pearcy AC, Boyan BD, Schwartz Z. Laryngeal Cancer Cells Metabolize 25-Hydroxyvitamin D 3 and Respond to 24R,25-dihydroxyvitamin D 3 via a Mechanism Dependent on Estrogen Receptor Levels. Cancers (Basel) 2024; 16:1635. [PMID: 38730587 PMCID: PMC11083835 DOI: 10.3390/cancers16091635] [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: 03/24/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Studies have evaluated vitamin D3's therapeutic potential in estrogen-responsive cancers, with conflicting findings. We have shown that the proliferation of breast cancer cells is regulated by 24R,25-dihydroxyvitamin D3 (24R,25(OH)2D3) depending on estrogen receptor alpha 66 (ERα66) expression, suggesting that this could also be the case for estrogen-sensitive laryngeal cancer cells. Accordingly, we examined levels of ERα isoforms in ERα66-positive UM-SCC-12 and ERα66-negative UM-SCC-11A cells and their response to 24R,25(OH)2D3. 24R,25(OH)2D3 stimulated proliferation, increased the expression of metastatic markers, and inhibited apoptosis in UM-SCC-12 cells while having the opposite effect in UM-SCC-11A cells. To evaluate if vitamin metabolites could act via autocrine/paracrine mechanisms, we assessed the expression, protein levels, and activity of vitamin D3 hydroxylases CYP24A1 and CYP27B1. Both cell types expressed both mRNAs; but the levels of the enzymes and their activities were differentially regulated by estrogen. ERα66-negative UM-SCC-11A cells produced more 24,25(OH)2D3 than UM-SCC-12 cells, but comparable levels of 1,25(OH)2D3 when treated with 25(OH)D3 These results suggest that the regulation of vitamin D3 metabolism in laryngeal cancer cells is modulated by ERα66 expression, and support a role for 24R,25(OH)2D3 as an autocrine/paracrine regulator of laryngeal cancer. The local metabolism of 25(OH)D3 should be considered when determining the potential of vitamin D3 in laryngeal cancer.
Collapse
Affiliation(s)
- Cydney D. Dennis
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (C.D.D.); (J.T.D.); (P.H.P.); (D.J.C.); (B.D.B.)
| | - Jonathan T. Dillon
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (C.D.D.); (J.T.D.); (P.H.P.); (D.J.C.); (B.D.B.)
| | - Prit H. Patel
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (C.D.D.); (J.T.D.); (P.H.P.); (D.J.C.); (B.D.B.)
| | - David J. Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (C.D.D.); (J.T.D.); (P.H.P.); (D.J.C.); (B.D.B.)
| | - Matthew S. Halquist
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.S.H.); (A.C.P.)
- Bioanalytical Core Laboratory, Central Virginia Drug Abuse Research Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Adam C. Pearcy
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.S.H.); (A.C.P.)
| | - Barbara D. Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (C.D.D.); (J.T.D.); (P.H.P.); (D.J.C.); (B.D.B.)
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (C.D.D.); (J.T.D.); (P.H.P.); (D.J.C.); (B.D.B.)
- Department of Periodontics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
6
|
Capaci V, Kharrat F, Conti A, Salviati E, Basilicata MG, Campiglia P, Balasan N, Licastro D, Caponnetto F, Beltrami AP, Monasta L, Romano F, Di Lorenzo G, Ricci G, Ura B. The Deep Proteomics Approach Identified Extracellular Vesicular Proteins Correlated to Extracellular Matrix in Type One and Two Endometrial Cancer. Int J Mol Sci 2024; 25:4650. [PMID: 38731868 PMCID: PMC11083465 DOI: 10.3390/ijms25094650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Among gynecological cancers, endometrial cancer is the most common in developed countries. Extracellular vesicles (EVs) are cell-derived membrane-surrounded vesicles that contain proteins involved in immune response and apoptosis. A deep proteomic approach can help to identify dysregulated extracellular matrix (ECM) proteins in EVs correlated to key pathways for tumor development. In this study, we used a proteomics approach correlating the two acquisitions-data-dependent acquisition (DDA) and data-independent acquisition (DIA)-on EVs from the conditioned medium of four cell lines identifying 428 ECM proteins. After protein quantification and statistical analysis, we found significant changes in the abundance (p < 0.05) of 67 proteins. Our bioinformatic analysis identified 26 pathways associated with the ECM. Western blotting analysis on 13 patients with type 1 and type 2 EC and 13 endometrial samples confirmed an altered abundance of MMP2. Our proteomics analysis identified the dysregulated ECM proteins involved in cancer growth. Our data can open the path to other studies for understanding the interaction among cancer cells and the rearrangement of the ECM.
Collapse
Affiliation(s)
- Valeria Capaci
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy (F.K.); (A.C.); (N.B.); (F.R.); (G.D.L.); (G.R.); (B.U.)
| | - Feras Kharrat
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy (F.K.); (A.C.); (N.B.); (F.R.); (G.D.L.); (G.R.); (B.U.)
| | - Andrea Conti
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy (F.K.); (A.C.); (N.B.); (F.R.); (G.D.L.); (G.R.); (B.U.)
| | - Emanuela Salviati
- Department of Pharmacy, University of Salerno, 84084 Salerno, Italy; (E.S.); (P.C.)
| | - Manuela Giovanna Basilicata
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Salerno, Italy; (E.S.); (P.C.)
| | - Nour Balasan
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy (F.K.); (A.C.); (N.B.); (F.R.); (G.D.L.); (G.R.); (B.U.)
| | | | - Federica Caponnetto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (F.C.); (A.P.B.)
| | - Antonio Paolo Beltrami
- Department of Medicine, University of Udine, 33100 Udine, Italy; (F.C.); (A.P.B.)
- Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Lorenzo Monasta
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy (F.K.); (A.C.); (N.B.); (F.R.); (G.D.L.); (G.R.); (B.U.)
| | - Federico Romano
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy (F.K.); (A.C.); (N.B.); (F.R.); (G.D.L.); (G.R.); (B.U.)
| | - Giovanni Di Lorenzo
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy (F.K.); (A.C.); (N.B.); (F.R.); (G.D.L.); (G.R.); (B.U.)
| | - Giuseppe Ricci
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy (F.K.); (A.C.); (N.B.); (F.R.); (G.D.L.); (G.R.); (B.U.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Blendi Ura
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy (F.K.); (A.C.); (N.B.); (F.R.); (G.D.L.); (G.R.); (B.U.)
| |
Collapse
|
7
|
Feng Y, Huo Q, Li BY, Yokota H. Unveiling the Dichotomy of Urinary Proteins: Diagnostic Insights into Breast and Prostate Cancer and Their Roles. Proteomes 2023; 12:1. [PMID: 38250812 PMCID: PMC10801584 DOI: 10.3390/proteomes12010001] [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: 11/10/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
This review covers the diagnostic potential of urinary biomarkers, shedding light on their linkage to cancer progression. Urinary biomarkers offer non-invasive avenues for detecting cancers, potentially bypassing the invasiveness of biopsies. The investigation focuses primarily on breast and prostate cancers due to their prevalence among women and men, respectively. The intricate interplay of urinary proteins is explored, revealing a landscape where proteins exhibit context-dependent behaviors. The review highlights the potential impact of physical activity on urinary proteins, suggesting its influence on tumorigenic behaviors. Exercise-conditioned urine may emerge as a potential diagnostic biomarker source. Furthermore, treatment effects, notably after lumpectomy and prostatectomy, induce shifts in the urinary proteome, indicating therapeutic impacts rather than activating oncogenic signaling. The review suggests further investigations into the double-sided, context-dependent nature of urinary proteins, the potential role of post-translational modifications (PTM), and the integration of non-protein markers like mRNA and metabolites. It also discusses a linkage of urinary proteomes with secretomes from induced tumor-suppressing cells (iTSCs). Despite challenges like cancer heterogeneity and sample variability due to age, diet, and comorbidities, harnessing urinary proteins and proteoforms may hold promise for advancing our understanding of cancer progressions, as well as the diagnostic and therapeutic role of urinary proteins.
Collapse
Affiliation(s)
- Yan Feng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China;
| | - Qingji Huo
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China;
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Bai-Yan Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China;
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| |
Collapse
|
8
|
Yan XR, Shi T, Xiao JY, Liu YF, Zheng HL. In vitro transdifferentiated signatures of goat preadipocytes into mammary epithelial cells revealed by DNA methylation and transcriptome profiling. J Biol Chem 2022; 298:102604. [PMID: 36257406 PMCID: PMC9668736 DOI: 10.1016/j.jbc.2022.102604] [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: 06/07/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
During mammary development, the transdifferentiation of mammary preadipocytes is one of the important sources for lactating mammary epithelial cells (MECs). However, there is limited knowledge about the mechanisms of dynamic regulation of transcriptome and genome-wide DNA methylation in the preadipocyte transdifferentiation process. Here, to gain more insight into these mechanisms, preadipocytes were isolated from adipose tissues from around the goat mammary gland (GM-preadipocytes). The GM-preadipocytes were cultured on Matrigel in conditioned media made from goat MECs to induce GM-preadipocyte-to-MEC transdifferentiation. The transdifferentiated GM-preadipocytes showed high abundance of keratin 18, which is a marker protein of MECs, and formed mammary acinar-like structures after 8 days of induction. Then, we performed transcriptome and DNA methylome profiling of the GM-preadipocytes and transdifferentiated GM-preadipocytes, respectively, and the differentially expressed genes and differentially methylated genes that play underlying roles in the process of transdifferentiation were obtained. Subsequently, we identified the candidate transcription factors in regulating the GM-preadipocyte-to-MEC transdifferentiation by transcription factor-binding motif enrichment analysis of differentially expressed genes and differentially methylated genes. Meanwhile, the secretory proteome of GM-preadipocytes cultured in conditioned media was also detected. By integrating the transcriptome, DNA methylome, and proteome, three candidate genes, four proteins, and several epigenetic regulatory axes were further identified, which are involved in regulation of the cell cycle, cell polarity establishment, cell adhesion, cell reprogramming, and adipocyte plasticity. These findings provide novel insights into the molecular mechanism of preadipocyte transdifferentiation and mammary development.
Collapse
|
9
|
Cheng P, Cao T, Zhao X, Lu W, Miao S, Ning F, Wang D, Gao Y, Wang L, Pei G, Yang L. Nidogen1-enriched extracellular vesicles accelerate angiogenesis and bone regeneration by targeting Myosin-10 to regulate endothelial cell adhesion. Bioact Mater 2022; 12:185-197. [PMID: 35310379 PMCID: PMC8897190 DOI: 10.1016/j.bioactmat.2021.10.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/17/2022] Open
Abstract
The technique bottleneck of repairing large bone defects with tissue engineered bone is the vascularization of tissue engineered grafts. Although some studies have shown that extracellular vesicles (EVs) derived from bone marrow mesenchymal stem cells (BMSCs) promote bone healing and repair by accelerating angiogenesis, the effector molecules and the mechanism remain unclear, which fail to provide ideas for the future research and development of cell-free interventions. Here, we found that Nidogen1-enriched EV (EV-NID1) derived from BMSCs interferes with the formation and assembly of focal adhesions (FAs) by targeting myosin-10, thereby reducing the adhesion strength of rat arterial endothelial cells (RAECs) to the extracellular matrix (ECM), and enhancing the migration and angiogenesis potential of RAECs. Moreover, by delivery with composite hydrogel, EV-NID1 is demonstrated to promote angiogenesis and bone regeneration in rat femoral defects. This study identifies the intracellular binding target of EV-NID1 and further elucidates a novel approach and mechanism, thereby providing a cell-free construction strategy with precise targets for the development of vascularized tissue engineering products. Nidogen1 is enriched in extracellular vesicles (EV-NID1) derived from BMSCs. EV-NID1 interferes with the formation and assembly of focal adhesions (FAs). Myosin-10 was identified as the intracellular binding target of EV-NID1. The composite hydrogel loaded with EV-NID1 promotes the repair of bone defects by accelerating angiogenesis.
Collapse
Affiliation(s)
- Pengzhen Cheng
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Tianqing Cao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xueyi Zhao
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Weiguang Lu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Sheng Miao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Fenru Ning
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Dong Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Gao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Long Wang
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Guoxian Pei
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Corresponding author.
| | - Liu Yang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Corresponding author.
| |
Collapse
|
10
|
Proteomic Analysis of Lung Cancer Types—A Pilot Study. Cancers (Basel) 2022; 14:cancers14112629. [PMID: 35681609 PMCID: PMC9179298 DOI: 10.3390/cancers14112629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the leading cause of tumor-related mortality, therefore significant effort is directed towards understanding molecular alterations occurring at the origin of the disease to improve current treatment options. The aim of our pilot-scale study was to carry out a detailed proteomic analysis of formalin-fixed paraffin-embedded tissue sections from patients with small cell or non-small cell lung cancer (adenocarcinoma, squamous cell carcinoma, and large cell carcinoma). Tissue surface digestion was performed on relatively small cancerous and tumor-adjacent normal regions and differentially expressed proteins were identified using label-free quantitative mass spectrometry and subsequent statistical analysis. Principal component analysis clearly distinguished cancerous and cancer adjacent normal samples, while the four lung cancer types investigated had distinct molecular profiles and gene set enrichment analysis revealed specific dysregulated biological processes as well. Furthermore, proteins with altered expression unique to a specific lung cancer type were identified and could be the targets of future studies.
Collapse
|
11
|
Lepucki A, Orlińska K, Mielczarek-Palacz A, Kabut J, Olczyk P, Komosińska-Vassev K. The Role of Extracellular Matrix Proteins in Breast Cancer. J Clin Med 2022; 11:jcm11051250. [PMID: 35268340 PMCID: PMC8911242 DOI: 10.3390/jcm11051250] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/16/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix is a structure composed of many molecules, including fibrillar (types I, II, III, V, XI, XXIV, XXVII) and non-fibrillar collagens (mainly basement membrane collagens: types IV, VIII, X), non-collagenous glycoproteins (elastin, laminin, fibronectin, thrombospondin, tenascin, osteopontin, osteonectin, entactin, periostin) embedded in a gel of negatively charged water-retaining glycosaminoglycans (GAGs) such as non-sulfated hyaluronic acid (HA) and sulfated GAGs which are linked to a core protein to form proteoglycans (PGs). This highly dynamic molecular network provides critical biochemical and biomechanical cues that mediate the cell–cell and cell–matrix interactions, influence cell growth, migration and differentiation and serve as a reservoir of cytokines and growth factors’ action. The breakdown of normal ECM and its replacement with tumor ECM modulate the tumor microenvironment (TME) composition and is an essential part of tumorigenesis and metastasis, acting as key driver for malignant progression. Abnormal ECM also deregulate behavior of stromal cells as well as facilitating tumor-associated angiogenesis and inflammation. Thus, the tumor matrix modulates each of the classically defined hallmarks of cancer promoting the growth, survival and invasion of the cancer. Moreover, various ECM-derived components modulate the immune response affecting T cells, tumor-associated macrophages (TAM), dendritic cells and cancer-associated fibroblasts (CAF). This review article considers the role that extracellular matrix play in breast cancer. Determining the detailed connections between the ECM and cellular processes has helped to identify novel disease markers and therapeutic targets.
Collapse
Affiliation(s)
- Arkadiusz Lepucki
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
| | - Kinga Orlińska
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.M.-P.); (J.K.)
| | - Jacek Kabut
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.M.-P.); (J.K.)
| | - Pawel Olczyk
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
- Correspondence:
| | - Katarzyna Komosińska-Vassev
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland;
| |
Collapse
|
12
|
Zhang Y, Zhang N, Liu L, Wang Y, Xing J, Li X. Transcriptome Analysis of Effects of Folic Acid Supplement on Gene Expression in Liver of Broiler Chickens. Front Vet Sci 2021; 8:686609. [PMID: 34604366 PMCID: PMC8481781 DOI: 10.3389/fvets.2021.686609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/23/2021] [Indexed: 12/30/2022] Open
Abstract
Folic acid is a water-soluble B vitamin, and plays an important role in regulating gene expression and methylation. The liver is the major site of lipid biosynthesis in the chicken. Nevertheless, how gene expression and regulatory networks are affected by folic acid in liver of broilers are poorly understood. This paper conducted the RNA-seq technology on the liver of broilers under folic acid challenge investigation. First, 405 differentially expressed genes (DEGs), including 157 significantly upregulated and 248 downregulated, were detected between the control group (C) and the 5 mg folic acid group (M). Second, 68 upregulated DEGs and 142 downregulated DEGs were determined between C group and 10 mg folic acid group (H). Third, there were 165 upregulated genes and 179 downregulated genes between M and H groups. Of these DEGs, 903 DEGs were successfully annotated in the public databases. The functional classification based on GO and KEEGG showed that “general function prediction only” represented the largest functional classes, “cell cycle” (C vs. M; M vs. H), and “neuroactive ligand-receptor interaction” (C vs. H) were the highest unique sequences among three groups. SNP analysis indicated that numbers of C, M and H groups were 145,450, 146,131, and 123,004, respectively. Total new predicted alternative splicing events in C, M, and H groups were 9,521, 9,328, and 8,929, respectively. A protein-protein interaction (PPI) network was constructed, and the top 10 hub genes were evaluated among three groups. The results of real time PCR indicated that mRNA abundance of PPARγ and FAS in abdominal fat of M and H groups were reduced compared with the C group (P < 0.05). Ultramicroscopy results showed that folic acid could reduce lipid droplets in livers from chickens. Finally, contents of LPL, PPARγ, and FAS in abdominal fat were decreased with the folic acid supplmented diets (P < 0.01). These findings reveal the effects of folic acid supplemention on gene expression in liver of broilers, which can provide information for understanding the molecular mechanisms of folic acid regulating liver lipid metabolism.
Collapse
Affiliation(s)
- Yujie Zhang
- School of Life Sciences, Linyi University, Linyi, China
| | - Ningbo Zhang
- School of Agriculture and Forestry Sciences, Linyi University, Linyi, China
| | - Lin Liu
- School of Pharmacy, Linyi University, Linyi, China
| | - Yan Wang
- School of Life Sciences, Linyi University, Linyi, China
| | - Jinyi Xing
- School of Life Sciences, Linyi University, Linyi, China
| | - Xiuling Li
- School of Life Sciences, Linyi University, Linyi, China
| |
Collapse
|