1
|
Kuratani A, Okamoto M, Kishida K, Okuzaki D, Sasai M, Sakaguchi S, Arase H, Yamamoto M. Platelet factor 4-induced T H1-T reg polarization suppresses antitumor immunity. Science 2024; 386:eadn8608. [PMID: 39571033 DOI: 10.1126/science.adn8608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 09/08/2024] [Indexed: 11/24/2024]
Abstract
The tumor microenvironment (TME) contains a number of immune-suppressive cells such as T helper 1-polarized regulatory T cells (TH1-Treg cells). However, little is known about the mechanism behind the abundant presence of TH1-Treg cells in the TME. We demonstrate that selective depletion of arginase I (Arg1)-expressing tumor-associated macrophages (Arg1+ TAMs) inhibits tumor growth and concurrently reduces the ratio of TH1-Treg cells in the TME. Arg1+ TAMs secrete the chemokine platelet factor 4 (PF4), which reinforces interferon-γ (IFN-γ)-induced Treg cell polarization into TH1-Treg cells in a manner dependent on CXCR3 and the IFN-γ receptor. Both genetic PF4 inactivation and PF4 neutralization hinder TH1-Treg cell accumulation in the TME and reduce tumor growth. Collectively, our study highlights the importance of Arg1+ TAM-produced PF4 for high TH1-Treg cell levels in the TME to suppress antitumor immunity.
Collapse
Affiliation(s)
- Ayumi Kuratani
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Masaaki Okamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Kazuki Kishida
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Osaka University, Suita, Osaka, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Center for Advances Modalities and Drug Delivery Systems, Osaka University, Suita, Osaka, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Hisashi Arase
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Center for Advances Modalities and Drug Delivery Systems, Osaka University, Suita, Osaka, Japan
- Department of Immunochemistry, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Center for Advances Modalities and Drug Delivery Systems, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
2
|
Smith YE, Wang G, Flynn CL, Madden SF, MacEneaney O, Cruz RGB, Richards CE, Jahns H, Brennan M, Cremona M, Hennessy BT, Sheehan K, Casucci A, Sani FA, Hudson L, Fay J, Vellanki SH, O’Flaherty S, Devocelle M, Hill ADK, Brennan K, Sukumar S, Hopkins AM. Functional Antagonism of Junctional Adhesion Molecule-A (JAM-A), Overexpressed in Breast Ductal Carcinoma In Situ (DCIS), Reduces HER2-Positive Tumor Progression. Cancers (Basel) 2022; 14:cancers14051303. [PMID: 35267611 PMCID: PMC8909510 DOI: 10.3390/cancers14051303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Specific drug targets for breast ductal carcinoma in situ (DCIS) remain elusive, despite increasing disease prevalence and burden to healthcare services. Estrogen receptor (ER)-negative HER2-positive DCIS, associated with the poorest patient prognosis, is in particular need of novel therapeutic avenues. This report provides the first evidence that a cell surface protein called JAM-A is upregulated on human DCIS patient tissues and can be readily targeted by a novel JAM-A-binding peptide inhibitor in separate in vivo models of DCIS. The anti-tumor efficacy and lack of systemic toxicity of this lead inhibitor, coupled with early indications of potential signaling pathways implicated, support the value of future studies investigating JAM-A as a novel drug target in DCIS patients. Abstract Breast ductal carcinoma in situ (DCIS) is clinically challenging, featuring high diagnosis rates and few targeted therapies. Expression/signaling from junctional adhesion molecule-A (JAM-A) has been linked to poor prognosis in invasive breast cancers, but its role in DCIS is unknown. Since progression from DCIS to invasive cancer has been linked with overexpression of the human epidermal growth factor receptor-2 (HER2), and JAM-A regulates HER2 expression, we evaluated JAM-A as a therapeutic target in DCIS. JAM-A expression was immunohistochemically assessed in patient DCIS tissues. A novel JAM-A antagonist (JBS2) was designed and tested alone/in combination with the HER2 kinase inhibitor lapatinib, using SUM-225 cells in vitro and in vivo as validated DCIS models. Murine tumors were proteomically analyzed. JAM-A expression was moderate/high in 96% of DCIS patient tissues, versus 23% of normal adjacent tissues. JBS2 bound to recombinant JAM-A, inhibiting cell viability in SUM-225 cells and a primary DCIS culture in vitro and in a chick embryo xenograft model. JBS2 reduced tumor progression in in vivo models of SUM-225 cells engrafted into mammary fat pads or directly injected into the mammary ducts of NOD-SCID mice. Preliminary proteomic analysis revealed alterations in angiogenic and apoptotic pathways. High JAM-A expression in aggressive DCIS lesions and their sensitivity to treatment by a novel JAM-A antagonist support the viability of testing JAM-A as a novel therapeutic target in DCIS.
Collapse
Affiliation(s)
- Yvonne E. Smith
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Guannan Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (G.W.); (S.S.)
| | - Ciara L. Flynn
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Stephen F. Madden
- Data Science Centre, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland;
| | - Owen MacEneaney
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (O.M.); (K.S.); (J.F.)
| | - Rodrigo G. B. Cruz
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Cathy E. Richards
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Hanne Jahns
- School of Veterinary Medicine, University College Dublin, Dublin 4, Ireland;
| | - Marian Brennan
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland;
| | - Mattia Cremona
- Department of Medical Oncology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (M.C.); (B.T.H.)
| | - Bryan T. Hennessy
- Department of Medical Oncology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (M.C.); (B.T.H.)
| | - Katherine Sheehan
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (O.M.); (K.S.); (J.F.)
| | - Alexander Casucci
- School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (A.C.); (F.A.S.)
| | - Faizah A. Sani
- School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (A.C.); (F.A.S.)
| | - Lance Hudson
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Joanna Fay
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (O.M.); (K.S.); (J.F.)
| | - Sri H. Vellanki
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Siobhan O’Flaherty
- Department of Chemistry, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (S.O.); (M.D.)
| | - Marc Devocelle
- Department of Chemistry, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (S.O.); (M.D.)
| | - Arnold D. K. Hill
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Kieran Brennan
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (G.W.); (S.S.)
| | - Ann M. Hopkins
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
- Correspondence: ; Tel.: +353-1-809-3858
| |
Collapse
|
3
|
Lucia RM, Huang WL, Alvarez A, Masunaka I, Ziogas A, Goodman D, Odegaard AO, Norden-Krichmar TM, Park HL. Association of mammographic density with blood DNA methylation. Epigenetics 2021; 17:531-546. [PMID: 34116608 PMCID: PMC9067527 DOI: 10.1080/15592294.2021.1928994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background: Altered DNA methylation may be an intermediate phenotype between breast cancer risk factors and disease. Mammographic density is a strong risk factor for breast cancer. However, no studies to date have identified an epigenetic signature of mammographic density. We performed an epigenome-wide association study of mammographic density. Methods: White blood cell DNA methylation was measured for 385 postmenopausal women using the Illumina Infinium MethylationEPIC BeadChip array. Differential methylation was assessed using genome-wide, probe-level, and regional analyses. We implemented a resampling-based approach to improve the stability of our findings. Results: On average, women with elevated mammographic density exhibited DNA hypermethylation within CpG islands and gene promoters compared to women with lower mammographic density. We identified 250 CpG sites for which DNA methylation was significantly associated with mammographic density. The top sites were located within genes associated with cancer, including HDLBP, TGFB2, CCT4, and PAX8, and were more likely to be located in regulatory regions of the genome. We also identified differential DNA methylation in 37 regions, including within the promoters of PAX8 and PF4, a gene involved in the regulation of angiogenesis. Overall, our results paint a picture of epigenetic dysregulation associated with mammographic density. Conclusion: Mammographic density is associated with differential DNA methylation throughout the genome, including within genes associated with cancer. Our results suggest the potential involvement of several genes in the biological mechanisms behind differences in breast density between women. Further studies are warranted to explore these potential mechanisms and potential links to breast cancer risk.
Collapse
Affiliation(s)
- Rachel M Lucia
- Department of Epidemiology, University of California, Irvine, USA
| | - Wei-Lin Huang
- Department of Epidemiology, University of California, Irvine, USA
| | - Andrea Alvarez
- Department of Medicine, University of California, Irvine, USA
| | - Irene Masunaka
- Department of Medicine, University of California, Irvine, USA
| | - Argyrios Ziogas
- Department of Medicine, University of California, Irvine, USA
| | - Deborah Goodman
- Department of Epidemiology, University of California, Irvine, USA
| | | | | | - Hannah Lui Park
- Department of Epidemiology, University of California, Irvine, USA.,Department of Pathology and Laboratory Medicine, University of California, Irvine, USA
| |
Collapse
|
4
|
Wang J, Ma G, Li M, Han X, Xu J, Liang M, Mao X, Chen X, Xia T, Liu X, Wang S. Plasma tRNA Fragments Derived from 5' Ends as Novel Diagnostic Biomarkers for Early-Stage Breast Cancer. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 21:954-964. [PMID: 32814252 PMCID: PMC7452045 DOI: 10.1016/j.omtn.2020.07.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 06/15/2020] [Accepted: 07/20/2020] [Indexed: 01/11/2023]
Abstract
Small RNAs derived from tRNAs are attracting considerable attention; however, the effects of tRNA-derived fragments (tRFs) and tRNA halves (tiRNAs) as biomarkers have not been investigated in early-stage breast cancer (EBC). The study aimed to explore whether tRFs and tiRNAs could be detected in plasma and whether they could serve as diagnostic biomarkers. The study was conducted in four phases. Thirty tRFs and tiRNAs were selected by high-throughput sequencing in screening phase and then assessed in training, testing, and external validation phases by qRT-PCR. Six tRFs (tRF-Glu-CTC-003, tRF-Gly-CCC-007, tRF-Gly-CCC-008, tRF-Leu-CAA-003, tRF-Ser-TGA-001, and tRF-Ser-TGA-002) were found significantly downregulated in plasma samples of patients with EBC compared with normal controls, and all were derived from 5' ends of tRNAs. Patients with HER2+ EBC with low expression levels of tRF-Glu-CTC-003 were related to worse disease-free survival and overall survival. The identified tRFs were further examined in cell supernatants, exosomes isolated from plasma, and tissues. In conclusion, our study identified six tRFs from the 5' ends of tRNAs as novel diagnostic biomarkers for EBC, providing additional evidence for, and a better understanding of, circulating tRFs and EBC.
Collapse
Affiliation(s)
- Jingyi Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| | - Ge Ma
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| | - Minghui Li
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| | - Xu Han
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, China
| | - Jin Xu
- Department of Breast and Thyroid Surgery, Nanjing First Hospital, Nanjing Medical University, 210029 Nanjing, China
| | - Mengdi Liang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, China
| | - Xinrui Mao
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, China
| | - Xiang Chen
- Thyroid and Breast Department, Yixing People's Hospital affiliated to Jiangsu University, 214200 Yixing, China
| | - Tiansong Xia
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 211166 Nanjing, China.
| | - Xiaoan Liu
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 211166 Nanjing, China.
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029 Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 211166 Nanjing, China.
| |
Collapse
|
5
|
Bauer D, Mazzio E, Soliman KFA. Whole Transcriptomic Analysis of Apigenin on TNFα Immuno-activated MDA-MB-231 Breast Cancer Cells. Cancer Genomics Proteomics 2020; 16:421-431. [PMID: 31659097 DOI: 10.21873/cgp.20146] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/30/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer is categorized by a lack of hormone receptors, inefficacy of anti-estrogen or aromatase inhibitor chemotherapies and greater mortality rates in African American populations. Advanced-stage breast tumors have a high concentration of tumor necrosis factor-α (TNFα) throughout the tumor/stroma milieu, prompting sustained release of diverse chemokines (i.e. C-C motif chemokine ligand 2 (CCL2)/CCL5). These potent chemokines can subsequently direct mass infiltration of leukocyte sub-populations to lodge within the tumor, triggering a loss of tumor immune surveillance and subsequent rapid tumor growth. Previously, we demonstrated that in the MDA-MB-231 TNBC cell line, TNFα evoked a rise in immune signaling proteins: CCL2, granulocyte macrophage colony-stimulating factor, interleukin (IL)1α, IL6 and inhibitor of nuclear factor kappa-B kinase subunit epsilon (IKBKε) all of which were attenuated by apigenin, a dietary flavonoid found in chamomile and parsley. MATERIALS AND METHODS The present work elucidates changes evoked by TNFα in the presence or absence of apigenin by examining the entire transcriptome for mRNA and long intergenic non-coding RNA with Affymetrix Hugene-2.1_ST human microarrays. Differential gene-expression analysis was conducted on 48,226 genes. RESULTS TNFα caused up-regulation of 75 genes and down-regulation of 10. Of these, apigenin effectively down-regulated 35 of the 75 genes which were up-regulated by TNFα. These findings confirm our previous work, specifically for the TNFα-evoked spike in IL1A vs. untreated controls [+21-fold change (FC), p<0.0001] being attenuated by apigenin in the presence of TNFa (-15 FC vs. TNFα, p<0.0001). Similar trends were seen for apigenin-mediated down-regulation of TNFα-up-regulated transcripts: IKBKE (TNFα: 4.55 FC vs. control, p<0.001; and TNFα plus apigenin: -4.92 FC, p<0.001), CCL2 (2.19 FC, p<0.002; and -2.12 FC, p<0.003), IL6 (3.25 FC, p<0.020; and -2.85 FC, p<0.043) and CSF2 (TNFα +6.04 FC, p<0.001; and -2.36 FC, p<0.007). In addition, these data further establish more than a 65% reduction by apigenin for the following transcripts which were also up-regulated by TNFα: cathepsin S (CTSS), complement C3 (C3), laminin subunit gamma 2 (LAMC2), (TLR2), toll-like receptor 2 G protein-coupled receptor class C group 5 member B (GPRC5B), contactin-associated protein 1 (CNTNAP1), claudin 1 (CLDN1), nuclear factor of activated T-cells 2 (NFATC2), C-X-C motif chemokine ligand 10 (CXCL10), CXCL11, interleukin 1 receptor-associated kinase 3 (IRAK3), nuclear receptor subfamily 3 group C member 2 (NR3C2), interleukin 32 (IL32), IL24, slit guidance ligand 2 (SLIT2), transmembrane protein 132A (TMEM132A), TMEM171, signal transducing adaptor family member 2 (STAP2), mixed lineage kinase domain-like pseudokinase (MLKL), kinase insert domain receptor (KDR), BMP-binding endothelial regulator (BMPER), and kelch-like family member 36 (KLHL36). CONCLUSION There is a possible therapeutic role for apigenin in down-regulating diverse genes associated with tumorigenic leukocyte sub-population infiltration by triple-negative breast cancer. The data have been deposited into the Gene Expression Omnibus for public analysis at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE120550.
Collapse
Affiliation(s)
- David Bauer
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A
| | - Elizabeth Mazzio
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A
| | - Karam F A Soliman
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A.
| |
Collapse
|
6
|
Ruytinx P, Proost P, Struyf S. CXCL4 and CXCL4L1 in cancer. Cytokine 2018; 109:65-71. [DOI: 10.1016/j.cyto.2018.02.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/16/2018] [Accepted: 02/20/2018] [Indexed: 02/07/2023]
|
7
|
Jian J, Pang Y, Yan HH, Min Y, Achyut BR, Hollander MC, Lin PC, Liang X, Yang L. Platelet factor 4 is produced by subsets of myeloid cells in premetastatic lung and inhibits tumor metastasis. Oncotarget 2018; 8:27725-27739. [PMID: 27223426 PMCID: PMC5438604 DOI: 10.18632/oncotarget.9486] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/01/2016] [Indexed: 02/05/2023] Open
Abstract
Bone marrow-derived myeloid cells can form a premetastatic niche and provide a tumor-promoting microenvironment. However, subsets of myeloid cells have also been reported to have anti-tumor properties. It is not clear whether there is a transition between anti- and pro- tumor function of these myeloid cells, and if so, what are the underlying molecular mechanisms. Here we report platelet factor 4 (PF4), or CXCL4, but not the other family members CXCL9, 10, and 11, was produced at higher levels in the normal lung and early stage premetastatic lungs but decreased in later stage lungs. PF4 was mostly produced by Ly6G+CD11b+ myeloid cell subset. Although the number of Ly6G+CD11b+ cells was increased in the premetastatic lungs, the expression level of PF4 in these cells was decreased during the metastatic progression. Deletion of PF4 (PF4 knockout or KO mice) led an increased metastasis suggesting an inhibitory function of PF4. There were two underlying mechanisms: decreased blood vessel integrity in the premetastatic lungs and increased production of hematopoietic stem/progenitor cells (HSCs) and myeloid derived suppressor cells (MDSCs) in tumor-bearing PF4 KO mice. In cancer patients, PF4 expression levels were negatively correlated with tumor stage and positively correlated with patient survival. Our studies suggest that PF4 is a critical anti-tumor factor in the premetastatic site. Our finding of PF4 function in the tumor host provides new insight to the mechanistic understanding of tumor metastasis.
Collapse
Affiliation(s)
- Jiang Jian
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA.,Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Yanli Pang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA.,Current address: Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, P. R. China
| | - H Hannah Yan
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yongfen Min
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, NIH, Frederick, MD, USA
| | - Bhagelu R Achyut
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA.,Tumor Angiogenesis Laboratory, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - M Christine Hollander
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - P Charles Lin
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, NIH, Frederick, MD, USA
| | - Xinhua Liang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Li Yang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
8
|
Ajona D, Ortiz-Espinosa S, Pio R. Complement anaphylatoxins C3a and C5a: Emerging roles in cancer progression and treatment. Semin Cell Dev Biol 2017; 85:153-163. [PMID: 29155219 DOI: 10.1016/j.semcdb.2017.11.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/07/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Recent insights into the role of complement anaphylatoxins C3a and C5a in cancer provide new opportunities for the development of innovative biomarkers and therapeutic strategies. These two complement activation products can maintain chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and increase the motility and metastatic potential of cancer cells. Still, the diverse heterogeneity of responses mediated by these peptides poses a challenge both to our understanding of the role played by these molecules in cancer progression and to the development of effective treatments. This review attempts to summarize the evidence surrounding the involvement of anaphylatoxins in the biological contexts associated with tumor progression. We also describe the recent developments that support the inhibition of anaphylatoxins, or their cognate receptors C3aR and C5aR1, as a treatment option for maximizing the clinical efficacy of current immunotherapies that target the PD-1/PD-L1 immune checkpoint.
Collapse
Affiliation(s)
- Daniel Ajona
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Sergio Ortiz-Espinosa
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Ruben Pio
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain.
| |
Collapse
|
9
|
Beretov J, Wasinger VC, Millar EKA, Schwartz P, Graham PH, Li Y. Proteomic Analysis of Urine to Identify Breast Cancer Biomarker Candidates Using a Label-Free LC-MS/MS Approach. PLoS One 2015; 10:e0141876. [PMID: 26544852 PMCID: PMC4636393 DOI: 10.1371/journal.pone.0141876] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 10/14/2015] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Breast cancer is a complex heterogeneous disease and is a leading cause of death in women. Early diagnosis and monitoring progression of breast cancer are important for improving prognosis. The aim of this study was to identify protein biomarkers in urine for early screening detection and monitoring invasive breast cancer progression. METHOD We performed a comparative proteomic analysis using ion count relative quantification label free LC-MS/MS analysis of urine from breast cancer patients (n = 20) and healthy control women (n = 20). RESULTS Unbiased label free LC-MS/MS-based proteomics was used to provide a profile of abundant proteins in the biological system of breast cancer patients. Data analysis revealed 59 urinary proteins that were significantly different in breast cancer patients compared to the normal control subjects (p<0.05, fold change >3). Thirty-six urinary proteins were exclusively found in specific breast cancer stages, with 24 increasing and 12 decreasing in their abundance. Amongst the 59 significant urinary proteins identified, a list of 13 novel up-regulated proteins were revealed that may be used to detect breast cancer. These include stage specific markers associated with pre-invasive breast cancer in the ductal carcinoma in-situ (DCIS) samples (Leucine LRC36, MAST4 and Uncharacterized protein CI131), early invasive breast cancer (DYH8, HBA, PEPA, uncharacterized protein C4orf14 (CD014), filaggrin and MMRN2) and metastatic breast cancer (AGRIN, NEGR1, FIBA and Keratin KIC10). Preliminary validation of 3 potential markers (ECM1, MAST4 and filaggrin) identified was performed in breast cancer cell lines by Western blotting. One potential marker MAST4 was further validated in human breast cancer tissues as well as individual human breast cancer urine samples with immunohistochemistry and Western blotting, respectively. CONCLUSIONS Our results indicate that urine is a useful non-invasive source of biomarkers and the profile patterns (biomarkers) identified, have potential for clinical use in the detection of BC. Validation with a larger independent cohort of patients is required in the following study.
Collapse
Affiliation(s)
- Julia Beretov
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Kensington, Australia
- SEALS, Anatomical Pathology, St George Hospital, Kogarah, Australia
| | - Valerie C. Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW, Kensington, Australia
- School of Medical Sciences, UNSW, Kensington, Australia
| | - Ewan K. A. Millar
- SEALS, Anatomical Pathology, St George Hospital, Kogarah, Australia
- School of Medical Sciences, UNSW, Kensington, Australia
- Cancer Research Program, Kinghorn Cancer Centre and Garvan Institute of Medical Research, Darlinghurst, Australia
- School of Medicine and Health Sciences, University of Western Sydney, Campbelltown, Australia
| | - Peter Schwartz
- Breast Surgery, St George Private Hospital, Kogarah, Australia
| | - Peter H. Graham
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Kensington, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Kensington, Australia
- * E-mail:
| |
Collapse
|
10
|
Chung L, Moore K, Phillips L, Boyle FM, Marsh DJ, Baxter RC. Novel serum protein biomarker panel revealed by mass spectrometry and its prognostic value in breast cancer. Breast Cancer Res 2014; 16:R63. [PMID: 24935269 PMCID: PMC4095593 DOI: 10.1186/bcr3676] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 06/02/2014] [Indexed: 12/15/2022] Open
Abstract
Introduction Serum profiling using proteomic techniques has great potential to detect biomarkers that might improve diagnosis and predict outcome for breast cancer patients (BC). This study used surface-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) mass spectrometry (MS) to identify differentially expressed proteins in sera from BC and healthy volunteers (HV), with the goal of developing a new prognostic biomarker panel. Methods Training set serum samples from 99 BC and 51 HV subjects were applied to four adsorptive chip surfaces (anion-exchange, cation-exchange, hydrophobic, and metal affinity) and analyzed by time-of-flight MS. For validation, 100 independent BC serum samples and 70 HV samples were analyzed similarly. Cluster analysis of protein spectra was performed to identify protein patterns related to BC and HV groups. Univariate and multivariate statistical analyses were used to develop a protein panel to distinguish breast cancer sera from healthy sera, and its prognostic potential was evaluated. Results From 51 protein peaks that were significantly up- or downregulated in BC patients by univariate analysis, binary logistic regression yielded five protein peaks that together classified BC and HV with a receiver operating characteristic (ROC) area-under-the-curve value of 0.961. Validation on an independent patient cohort confirmed the five-protein parameter (ROC value 0.939). The five-protein parameter showed positive association with large tumor size (P = 0.018) and lymph node involvement (P = 0.016). By matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS, immunoprecipitation and western blotting the proteins were identified as a fragment of apolipoprotein H (ApoH), ApoCI, complement C3a, transthyretin, and ApoAI. Kaplan-Meier analysis on 181 subjects after median follow-up of >5 years demonstrated that the panel significantly predicted disease-free survival (P = 0.005), its efficacy apparently greater in women with estrogen receptor (ER)-negative tumors (n = 50, P = 0.003) compared to ER-positive (n = 131, P = 0.161), although the influence of ER status needs to be confirmed after longer follow-up. Conclusions Protein mass profiling by MS has revealed five serum proteins which, in combination, can distinguish between serum from women with breast cancer and healthy control subjects with high sensitivity and specificity. The five-protein panel significantly predicts recurrence-free survival in women with ER-negative tumors and may have value in the management of these patients.
Collapse
|
11
|
Han MY, Dai JJ, Zhang Y, Lin Q, Jiang M, Xu XY, Liu Q. Identification of osteoarthritis biomarkers by proteomic analysis of synovial fluid. J Int Med Res 2013; 40:2243-50. [PMID: 23321181 DOI: 10.1177/030006051204000622] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To use proteomic analysis to identify novel candidate biomarker proteins in synovial fluid for the differential diagnosis of osteoarthritis and rheumatoid arthritis. METHODS Synovial fluid samples were analysed using surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF-MS). Data were used to generate an artificial neural network (ANN). The identification of one protein peak was confirmed via Western blotting. RESULTS Fluid samples were analysed from 36 patients with osteoarthritis and 24 with rheumatoid arthritis. In total, three protein peaks (mass-to-charge ratio [m/z] 3893, 10,576 and 14,175 Da) were identified as potential biomarkers for osteoarthritis. The ANN differentiated between osteoarthritis and rheumatoid arthritis with a sensitivity of 89.4% and a specificity of 91.2%. The protein peak at m/z 10 576 was identified as S100 calcium binding protein A12 (S100A12). CONCLUSIONS A combination of SELDI-TOF-MS and ANN identified osteoarthritis biomarkers. SELDI-TOF-MS may be a useful tool in the screening of synovial fluid for osteoarthritis diagnosis.
Collapse
Affiliation(s)
- M Y Han
- Cancer Therapy and Research Centre, Shandong Provincial Hospital, Shandong University, Jinan Province, China
| | | | | | | | | | | | | |
Collapse
|
12
|
Grandjean M, Dieu M, Raes M, Feron O. A new method combining sequential immunoaffinity depletion and differential in gel electrophoresis to identify autoantibodies as cancer biomarkers. J Immunol Methods 2013; 396:23-32. [PMID: 23916966 DOI: 10.1016/j.jim.2013.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 12/17/2022]
Abstract
Easily measurable biomarkers are urgently required to detect early stages of cancer progression. Autoantibodies (aAbs), as a component of the humoral immune response against tumor cells, have such potential of diagnostic markers since they are circulating and stable proteins, produced rapidly and easily amenable to in vitro dosage. The identification of aAbs is based on the characterization of tumor-associated antigens (TAA) against which they are directed. Here, we propose a new method for an unbiased identification of TAA and thereby of aAbs as cancer biomarkers. This method that we called sequential immunoaffinity depletion-differential in gel electrophoresis (SID-DIGE) is based on the immunodepletion of tumor cell lysates with IgG from control and tumor-bearing mice and direct matching of the flow throughs of these immunoaffinity separations on the same 2D format. This strategy reduces the complexity of the samples to be analyzed and maximizes the interest of assessing hundreds of proteins simultaneously. SID-DIGE has also the potential, contrary to existing serological proteome analysis (SERPA) techniques, to detect immunogenic proteins with conformational epitopes, including those resulting from post-translational modifications. Using a model of human colorectal tumors in mice for the proof of principle, we showed that SID-DIGE outperforms the conventional SERPA technique, with the identification of 7 common TAA (validating our approach) and 18 additional aAbs proving the potential of this new method. In particular, the identification of aAbs directed against key enzymes supporting glycolysis gives credential to the role of hypoxia as a major determinant of the tumor proteome and thus as a source of immunogenicity. Overall, the developed methodology allowed efficient screening of sera for the identification of aAbs as potential biomarkers.
Collapse
Affiliation(s)
- Marie Grandjean
- UCLouvain, Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Brussels, Belgium
| | | | | | | |
Collapse
|
13
|
Klos A, Wende E, Wareham KJ, Monk PN. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXVII. Complement peptide C5a, C4a, and C3a receptors. Pharmacol Rev 2013; 65:500-43. [PMID: 23383423 DOI: 10.1124/pr.111.005223] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The activation of the complement cascade, a cornerstone of the innate immune response, produces a number of small (74-77 amino acid) fragments, originally termed anaphylatoxins, that are potent chemoattractants and secretagogues that act on a wide variety of cell types. These fragments, C5a, C4a, and C3a, participate at all levels of the immune response and are also involved in other processes such as neural development and organ regeneration. Their primary function, however, is in inflammation, so they are important targets for the development of antiinflammatory therapies. Only three receptors for complement peptides have been found, but there are no satisfactory antagonists as yet, despite intensive investigation. In humans, there is a single receptor for C3a (C3a receptor), no known receptor for C4a, and two receptors for C5a (C5a₁ receptor and C5a₂ receptor). The most recently characterized receptor, the C5a₂ receptor (previously known as C5L2 or GPR77), has been regarded as a passive binding protein, but signaling activities are now ascribed to it, so we propose that it be formally identified as a receptor and be given a name to reflect this. Here, we describe the complex biology of the complement peptides, introduce a new suggested nomenclature, and review our current knowledge of receptor pharmacology.
Collapse
Affiliation(s)
- Andreas Klos
- Department for Medical Microbiology, Medical School Hannover, Hannover, Germany
| | | | | | | |
Collapse
|
14
|
Kapoor S. Platelet factor 4 and its modulatory effect on systemic carcinogenesis. Thromb Res 2013; 131:e49. [DOI: 10.1016/j.thromres.2012.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 10/26/2012] [Accepted: 10/30/2012] [Indexed: 11/29/2022]
|
15
|
MALDI-MS-Based Profiling of Serum Proteome: Detection of Changes Related to Progression of Cancer and Response to Anticancer Treatment. INTERNATIONAL JOURNAL OF PROTEOMICS 2012; 2012:926427. [PMID: 22900176 PMCID: PMC3413974 DOI: 10.1155/2012/926427] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/12/2012] [Accepted: 06/12/2012] [Indexed: 01/25/2023]
Abstract
Mass spectrometry-based analyses of the low-molecular-weight fraction of serum proteome allow identifying proteome profiles (signatures) that are potentially useful in detection and classification of cancer. Several published studies have shown that multipeptide signatures selected in numerical tests have potential values for diagnostics of different types of cancer. However due to apparent problems with standardization of methodological details, both experimental and computational, none of the proposed peptide signatures analyzed directly by MALDI/SELDI-ToF spectrometry has been approved for routine diagnostics. Noteworthy, several components of proposed cancer signatures, especially those characteristic for advanced cancer, were identified as fragments of blood proteins involved in the acute phase and inflammatory response. This indicated that among cancer biomarker candidates to be possibly identified by serum proteome profiling were rather those reflecting overall influence of a disease (and the therapy) upon the human organism, than products of cancer-specific genes. Current paper focuses on changes in serum proteome that are related to response of patient's organism to progressing malignancy and toxicity of anticancer treatment. In addition, several methodological issues that affect robustness and interlaboratory reproducibility of MS-based serum proteome profiling are discussed.
Collapse
|
16
|
Selvaraju S, El Rassi Z. Tandem lectin affinity chromatography monolithic columns with surface immobilised concanavalin A, wheat germ agglutinin and Ricinus communis
agglutinin-I for capturing sub-glycoproteomics from breast cancer and disease-free human sera. J Sep Sci 2012; 35:1785-95. [DOI: 10.1002/jssc.201200230] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Ziad El Rassi
- Department of Chemistry; Oklahoma State University; Stillwater OK USA
| |
Collapse
|
17
|
Death is associated with complement C3 depletion in cerebrospinal fluid of patients with pneumococcal meningitis. mBio 2012; 3:mBio.00272-11. [PMID: 22415003 PMCID: PMC3312214 DOI: 10.1128/mbio.00272-11] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pneumococcal meningitis can lead to death or serious neurological sequelae as a result of the host inflammatory response. We investigated the association between host response protein expression and outcome in patients with pneumococcal meningitis. Cerebrospinal fluid (CSF) was obtained from 80 patients with pneumococcal meningitis (40 nonsurvivors and 40 survivors) and 10 normal controls. Candidate proteins were analyzed for an association with survival. Complement C3 levels were 5-fold lower in nonsurvivors than in survivors (P < 0.05). This C3 reduction was not associated with lower levels in serum, indicating a compartmentalized CSF response. Transferrin levels were significantly higher in CSF (but not serum) from nonsurvivors than in CSF from survivors, suggestive of blood-brain barrier damage. Classical apoptosis proteins caspase 3 and apoptosis-inducing factor were not present in CSF. Expression of creatine kinase BB in clinically infected CSF suggested neuronal necrosis, but there was no clear association between level of expression and clinical outcome. Increased blood-brain barrier permeability and complement C3 depletion may have a role in determining outcome from bacterial meningitis. Therapeutic use of citicoline or caspase inhibitors is unlikely to have beneficial effects in patients with meningitis. We previously identified proteins associated with clinical outcome in patients diagnosed with pneumococcal meningitis in a pilot proteomics study of cerebrospinal fluid (CSF). In this article, we have quantitatively assayed specific proteins identified from this previous proteomics analysis along with proteins associated with cell death by using Western blotting.
Collapse
|
18
|
Early diagnostic protein biomarkers for breast cancer: how far have we come? Breast Cancer Res Treat 2011; 134:1-12. [PMID: 22179926 DOI: 10.1007/s10549-011-1907-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 11/29/2011] [Indexed: 12/22/2022]
Abstract
Many studies have used surface-enhanced laser desorption/ionization time-of-flight mass spectrometry or matrix-assisted laser desorption/ionization time-of-flight mass spectrometry to search for blood-based proteins that are related to the presence of breast cancer. We review the biomarkers discovered or targeted measured by these methods and discuss the strengths and weaknesses of these studies. We highlight two proteins that were most often related to breast cancer: C3a des-arginine anaphylatoxin (C3adesArg) (molecular weight: 8,938 Da) and fragments of inter-alpha trypsin inhibitor heavy chain H4 (ITIH4). In addition, we elaborate on three important methodological aspects related to these studies: protein identification, specificity of the markers, and disease heterogeneity. Finally, we propose some points to be addressed in future studies. These include the use of other analytical measurement techniques, need of protein identification, the importance of identical sample handling protocols for cases and controls, and the stratification of the results according to molecular subtypes and stages of breast cancer. Ultimately this may lead to the discovery of new and valid breast cancer specific biomarkers.
Collapse
|
19
|
Identification of Potential Markers Related to Neoadjuvant Chemotherapy Sensitivity of Breast Cancer by SELDI-TOF MS. Appl Biochem Biotechnol 2011; 166:753-63. [DOI: 10.1007/s12010-011-9464-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 11/15/2011] [Indexed: 11/27/2022]
|
20
|
Taguchi A, Politi K, Pitteri SJ, Lockwood WW, Faça VM, Kelly-Spratt K, Wong CH, Zhang Q, Chin A, Park KS, Goodman G, Gazdar AF, Sage J, Dinulescu DM, Kucherlapati R, DePinho RA, Kemp CJ, Varmus HE, Hanash SM. Lung cancer signatures in plasma based on proteome profiling of mouse tumor models. Cancer Cell 2011; 20:289-99. [PMID: 21907921 PMCID: PMC3406925 DOI: 10.1016/j.ccr.2011.08.007] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 07/06/2011] [Accepted: 08/05/2011] [Indexed: 12/23/2022]
Abstract
We investigated the potential of in-depth quantitative proteomics to reveal plasma protein signatures that reflect lung tumor biology. We compared plasma protein profiles of four mouse models of lung cancer with profiles of models of pancreatic, ovarian, colon, prostate, and breast cancer and two models of inflammation. A protein signature for Titf1/Nkx2-1, a known lineage-survival oncogene in lung cancer, was found in plasmas of mouse models of lung adenocarcinoma. An EGFR signature was found in plasma of an EGFR mutant model, and a distinct plasma signature related to neuroendocrine development was uncovered in the small-cell lung cancer model. We demonstrate relevance to human lung cancer of the protein signatures identified on the basis of mouse models.
Collapse
Affiliation(s)
- Ayumu Taguchi
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Katerina Politi
- Program in Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | - William W. Lockwood
- Program in Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Vitor M. Faça
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Chee-Hong Wong
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Qing Zhang
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Alice Chin
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Kwon-Sik Park
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Gary Goodman
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Adi F. Gazdar
- Hamon Center for Therapeutic Oncology Research and Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Julien Sage
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Daniela M. Dinulescu
- Eugene Braunwald Research Center, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Raju Kucherlapati
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Ronald A. DePinho
- Belfer Institute for Applied Cancer Science, Department of Medical Oncology, Department of Medicine and Department of Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | - Harold E. Varmus
- Program in Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Samir M. Hanash
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Correspondence:
| |
Collapse
|
21
|
Dudley E, Hässler F, Thome J. Profiling for novel proteomics biomarkers in neurodevelopmental disorders. Expert Rev Proteomics 2011; 8:127-36. [PMID: 21329432 DOI: 10.1586/epr.10.97] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Protein biomarker discovery from biological fluids, such as serum, has been widely applied to disorders such as cancer and has more recently also been utilized in neuro-psychiatric disorders with relatively clear biological causes, such as Alzheimer's disease and schizophrenia. The application of the associated technologies for the identification of protein biomarker signatures in neurodevelopmental disorders, such as autism spectrum disorder and attention deficit hyperactivity disorder, is comparatively less well established. The aim of this article is to provide an overview of the various protocols available for such analysis, discuss reports in which these techniques have been previously applied in biomarker discovery/validation in neurodevelopmental disorders, and consider the future development of this area of research.
Collapse
Affiliation(s)
- Ed Dudley
- Institute of Mass Spectrometry, School of Medicine, Swansea University, Swansea, UK
| | | | | |
Collapse
|
22
|
ARIMITSU J, HAGIHARA K, OTSUKA S, NAKANISHI M, KISHIDA Y, INOUE T, KATO Y, OTANI Y, OZAKI K, SHIMIZU K, KAGEYAMA M, NISHIDA S. The Effect of Saffron in Patients with Autoimmune Diseases. ACTA ACUST UNITED AC 2011. [DOI: 10.3937/kampomed.62.548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|