1
|
Kasoha M, Findeklee S, Nigdelis MP, Schmidt G, Solomayer EF, Haj Hamoud B. Retrospective Evaluation of Bone Turnover Markers in Serum for the Prediction of Metastases Development in Breast Cancer Patients: A Cohort Study. Biomedicines 2024; 12:1201. [PMID: 38927408 PMCID: PMC11201037 DOI: 10.3390/biomedicines12061201] [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: 04/16/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Serum bone turnover markers might play a role in the prediction of the development of bone metastases in breast cancer (BC) patients. We conducted a retrospective cohort study to address the association of serum bone turnover markers with oncologic outcomes. METHODS We included 80 women with BC, who were operated on at the Department of Gynecology, Obstetrics and Reproductive Medicine, Homburg/Saar, Germany. Serum samples were obtained prior to surgery and were used for estimation of the concentration of tumor and bone turnover markers using enzyme-linked immunosorbent assay (ELISA) and radioimmunoassay (RIA). RESULTS At baseline, pyridinoline cross-linked carboxy-terminal telopeptide of type-1 collagen (ICTP) concentrations were higher in nodal positive vs. negative tumors (Mann-Whitney test p = 0.04). After a median follow-up of 79.4 months, 17 patients developed metastases, with 9 demonstrating, among other organs, osseous metastases. ICTP demonstrated the best area under the curve in the predection of osseous metastases in our cohort (AUC = 0.740, DeLong Test p = 0.005). Univariable Cox proportional hazard models failed to demonstrate significant associations between serum bone turnover markers and oncologic outcomes (progression-free survival, overall survival). CONCLUSIONS Serum bone turnover markers (e.g., ICTP) were able to predict the development of osseous metastases but were not associated with oncologic outcomes. Further investigation and validation are required for the use of such markers in clinical practice.
Collapse
Affiliation(s)
- Mariz Kasoha
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, D-66421 Homburg, Germany; (S.F.); (M.P.N.); (G.S.); (E.-F.S.); (B.H.H.)
| | - Sebastian Findeklee
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, D-66421 Homburg, Germany; (S.F.); (M.P.N.); (G.S.); (E.-F.S.); (B.H.H.)
- Medizinische Versorgungszentrum, Göttingen, Kasseler Landstraße 25a, D-37081 Göttingen, Germany
| | - Meletios P. Nigdelis
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, D-66421 Homburg, Germany; (S.F.); (M.P.N.); (G.S.); (E.-F.S.); (B.H.H.)
| | - Gilda Schmidt
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, D-66421 Homburg, Germany; (S.F.); (M.P.N.); (G.S.); (E.-F.S.); (B.H.H.)
| | - Erich-Franz Solomayer
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, D-66421 Homburg, Germany; (S.F.); (M.P.N.); (G.S.); (E.-F.S.); (B.H.H.)
| | - Bashar Haj Hamoud
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, D-66421 Homburg, Germany; (S.F.); (M.P.N.); (G.S.); (E.-F.S.); (B.H.H.)
| |
Collapse
|
2
|
Kasoha M, Steinbach AK, Bohle RM, Linxweiler B, Haj Hamoud B, Doerk M, Nigdelis MP, Stotz L, Zimmermann JSM, Solomayer EF, Kaya AC, Radosa JC. Dkk1 as a Prognostic Marker for Neoadjuvant Chemotherapy Response in Breast Cancer Patients. Cancers (Basel) 2024; 16:419. [PMID: 38254908 PMCID: PMC10814026 DOI: 10.3390/cancers16020419] [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: 12/11/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
PURPOSE To investigate the role of Dkk1 as a predictor of response to NACT in BC patients. METHODS This retrospective monocentric study included 145 women who had undergone NACT followed by breast surgery. Dkk1 protein expression was assessed using immunohistochemistry staining in core needle biopsies and mammary carcinoma specimens. RESULTS Dkk1 levels were lower in treated BC tumours than in untreated tumours. The outcomes of 68 matched pre- and post-therapy tissues showed that Dkk1 levels in mammary carcinoma tissues were significantly predicted by levels in core needle biopsies and that Dkk1 expression was reduced in 83% of cases. Smaller cT stage, positive Her2 expression, and decreased Dkk1-IRS in core needle biopsy tissues were all independent predictors of regression grade (R4), according to Sinn. However, the percentage of Dkk1 expression differences prior to and following NACT had no effect on PFS or OS. CONCLUSIONS In this study, we demonstrated for the first time that Dkk1 could be identified as an independent predictor of NACT response in BC patients, particularly those with TNBC. Further research with a multicentric expanded (pre-/post-therapy) sample set and better-defined populations in terms of molecular subtypes, therapy modality, and long-term follow-up is recommended to obtain more solid evidence.
Collapse
Affiliation(s)
- Mariz Kasoha
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Anna K. Steinbach
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Rainer M. Bohle
- Institute of General and Surgical Pathology, University Medical School of Saarland, 66421 Homburg, Germany;
| | - Barbara Linxweiler
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Bashar Haj Hamoud
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Merle Doerk
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Meletios P. Nigdelis
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Lisa Stotz
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Julia S. M. Zimmermann
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Erich-Franz Solomayer
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Askin C. Kaya
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| | - Julia C. Radosa
- Department of Gynaecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, 66421 Homburg, Germany; (A.K.S.); (B.L.); (B.H.H.); (M.P.N.); (L.S.); (J.S.M.Z.); (E.-F.S.); (A.C.K.); (J.C.R.)
| |
Collapse
|
3
|
Zhang X, Zhang R, Hou C, He R, Wang QS, Zhou TH, Li XQ, Zhai QL, Feng YM. FOXF2 oppositely regulates stemness in luminal and basal-like breast cancer cells through the Wnt/beta-catenin pathway. J Biol Chem 2022; 298:102082. [PMID: 35660418 PMCID: PMC9254110 DOI: 10.1016/j.jbc.2022.102082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 10/25/2022] Open
Abstract
The stemness of cancer cells contributes to tumorigenesis, the heterogeneity of malignancies, cancer metastasis, and therapeutic resistance. However, the role and regulatory mechanisms maintaining stemness among breast cancer subtypes remain elusive. Our previous studies have demonstrated that ectopic expression and dynamic alteration of the mesenchymal transcription factor forkhead box F2 (FOXF2) differentially regulates breast cancer progression and metastasis organotropism in a cell subtype-specific manner. Here, we reveal the underlying mechanism by which FOXF2 enhances stemness in luminal breast cancer cells but suppresses that in basal-like breast cancer (BLBC) cells. We show that luminal breast cancer and BLBC cells with FOXF2-regulated stemness exhibit partial mesenchymal stem cell properties that toward osteogenic differentiation and myogenic differentiation, respectively. Furthermore, we show that FOXF2 activates the Wnt signaling pathway in luminal breast cancer cells but represses this pathway in BLBC cells by recruiting nuclear receptor coactivator 3 (NCoA3) and nuclear receptor corepressor 1 (NCoR1) to the promoters of Wnt family member 2B (WNT2B) and frizzled class receptor 1 (FZD1) genes to activate and repress their transcription, respectively. We propose that targeting the Wnt signaling pathway is a promising strategy for the treatment of breast cancers with dysregulated expression of FOXF2.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Rui Zhang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Chen Hou
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Rui He
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Qing-Shan Wang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Tian-Hao Zhou
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Xiao-Qing Li
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Qiong-Li Zhai
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Yu-Mei Feng
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China.
| |
Collapse
|
4
|
Dahlmann M, Monks A, Harris ED, Kobelt D, Osterland M, Khaireddine F, Herrmann P, Kemmner W, Burock S, Walther W, Shoemaker RH, Stein U. Combination of Wnt/β-Catenin Targets S100A4 and DKK1 Improves Prognosis of Human Colorectal Cancer. Cancers (Basel) 2021; 14:cancers14010037. [PMID: 35008201 PMCID: PMC8750436 DOI: 10.3390/cancers14010037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/16/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022] Open
Abstract
Metastasis is directly linked to colorectal cancer (CRC) patient survival. Wnt signaling through β-catenin plays a key role. Metastasis-inducing S100A4 is a Wnt/β-catenin target gene and a prognostic biomarker for CRC and other cancer types. We aimed to identify S100A4-dependent expression alterations to better understand CRC progression and metastasis for improved patient survival. S100A4-induced transcriptome arrays, confirmatory studies in isogenic CRC cell lines with defined β-catenin genotypes, and functional metastasis studies were performed. S100A4-regulated transcriptome examination revealed the transcriptional cross-regulation of metastasis-inducing S100A4 with Wnt pathway antagonist Dickkopf-1 (DKK1). S100A4 overexpression down-regulated DKK1, S100A4 knock-down increased DKK1. Recombinant DKK1 reduced S100A4 expression and S100A4-mediated cell migration. In xenografted mice, systemic S100A4-shRNA application increased intratumoral DKK1. The inverse correlation of S100A4 and DKK1 was confirmed in five independent publicly available CRC expression datasets. Combinatorial analysis of S100A4 and DKK1 in two additional independent CRC patient cohorts improved prognosis of overall and metastasis-free survival. The newly discovered transcriptional cross-regulation of Wnt target S100A4 and Wnt antagonist DKK1 is predominated by an S100A4-induced Wnt signaling feedback loop, increasing cell motility and metastasis risk. S100A4 and DKK1 combination improves the identification of CRC patients at high risk.
Collapse
Affiliation(s)
- Mathias Dahlmann
- Experimental and Clinical Research Center, a Cooperation between the Charité—Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany; (M.D.); (D.K.); (M.O.); (F.K.); (P.H.); (W.K.); (W.W.)
| | - Anne Monks
- Molecular Pharmacology Laboratory, Leidos Biomedical Research, Inc., FNLCR, Frederick, MD 21702, USA; (A.M.); (E.D.H.)
| | - Erik D. Harris
- Molecular Pharmacology Laboratory, Leidos Biomedical Research, Inc., FNLCR, Frederick, MD 21702, USA; (A.M.); (E.D.H.)
| | - Dennis Kobelt
- Experimental and Clinical Research Center, a Cooperation between the Charité—Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany; (M.D.); (D.K.); (M.O.); (F.K.); (P.H.); (W.K.); (W.W.)
| | - Marc Osterland
- Experimental and Clinical Research Center, a Cooperation between the Charité—Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany; (M.D.); (D.K.); (M.O.); (F.K.); (P.H.); (W.K.); (W.W.)
| | - Fadi Khaireddine
- Experimental and Clinical Research Center, a Cooperation between the Charité—Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany; (M.D.); (D.K.); (M.O.); (F.K.); (P.H.); (W.K.); (W.W.)
| | - Pia Herrmann
- Experimental and Clinical Research Center, a Cooperation between the Charité—Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany; (M.D.); (D.K.); (M.O.); (F.K.); (P.H.); (W.K.); (W.W.)
| | - Wolfgang Kemmner
- Experimental and Clinical Research Center, a Cooperation between the Charité—Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany; (M.D.); (D.K.); (M.O.); (F.K.); (P.H.); (W.K.); (W.W.)
| | - Susen Burock
- Charité Comprehensive Cancer Center, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, Invalidenstraße 80, 10117 Berlin, Germany;
| | - Wolfgang Walther
- Experimental and Clinical Research Center, a Cooperation between the Charité—Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany; (M.D.); (D.K.); (M.O.); (F.K.); (P.H.); (W.K.); (W.W.)
| | - Robert H. Shoemaker
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute-Frederick, Building 440, Frederick, MD 21702, USA;
| | - Ulrike Stein
- Experimental and Clinical Research Center, a Cooperation between the Charité—Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany; (M.D.); (D.K.); (M.O.); (F.K.); (P.H.); (W.K.); (W.W.)
- German Cancer Consortium, 69121 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
5
|
Rimal R, Desai P, Marquez AB, Sieg K, Marquardt Y, Singh S. 3-D vascularized breast cancer model to study the role of osteoblast in formation of a pre-metastatic niche. Sci Rep 2021; 11:21966. [PMID: 34754042 PMCID: PMC8578551 DOI: 10.1038/s41598-021-01513-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Breast cancer cells (BCCs) preferentially metastasize to bone. It is known that BCCs remotely primes the distant bone site prior to metastasis. However, the reciprocal influence of bone cells on the primary tumor is relatively overlooked. Here, to study the bone-tumor paracrine influence, a tri-cellular 3-D vascularized breast cancer tissue (VBCTs) model is engineered which comprised MDA-MB231, a triple-negative breast cancer cells (TNBC), fibroblasts, and endothelial cells. This is indirectly co-cultured with osteoblasts (OBs), thereby constituting a complex quad-cellular tumor progression model. VBCTs alone and in conjunction with OBs led to abnormal vasculature and reduced vessel density but enhanced VEGF production. A total of 1476 significantly upregulated and 775 downregulated genes are identified in the VBCTs exposed to OBs. HSP90N, CYCS, RPS27A, and EGFR are recognized as upregulated hub-genes. Kaplan Meier plot shows HSP90N to have a significant outcome in TNBC patient survivability. Furthermore, compared to cancer tissues without vessels, gene analysis recognized 1278 significantly upregulated and 566 downregulated genes in VBCTs. DKK1, CXCL13, C3 protein and BMP4 are identified to be downregulated hub genes in VBCTs. Together, a multi-cellular breast cancer model and culture protocols are established to study pre-metastatic events in the presence of OBs.
Collapse
Affiliation(s)
- Rahul Rimal
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany
| | - Prachi Desai
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany
| | - Andrea Bonnin Marquez
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany
| | - Karina Sieg
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany
| | - Yvonne Marquardt
- Department of Dermatology and Allergology, University Hospital, RWTH Aachen University, 52074, Aachen, Germany
| | - Smriti Singh
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany.
- Max Planck Institute for Medical Research, Jahnstrasse 29, 69120, Heidelberg, Germany.
| |
Collapse
|
6
|
A Novel Approach for the Discovery of Biomarkers of Radiotherapy Response in Breast Cancer. J Pers Med 2021; 11:jpm11080796. [PMID: 34442440 PMCID: PMC8399231 DOI: 10.3390/jpm11080796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 01/08/2023] Open
Abstract
Radiotherapy (RT) is an important treatment modality for the local control of breast cancer (BC). Unfortunately, not all patients that receive RT will obtain a therapeutic benefit, as cancer cells that either possess intrinsic radioresistance or develop resistance during treatment can reduce its efficacy. For RT treatment regimens to become personalised, there is a need to identify biomarkers that can predict and/or monitor a tumour's response to radiation. Here we describe a novel method to identify such biomarkers. Liquid chromatography-mass spectrometry (LC-MS) was used on conditioned media (CM) samples from a radiosensitive oestrogen receptor positive (ER+) BC cell line (MCF-7) to identify cancer-secreted biomarkers which reflected a response to radiation. A total of 33 radiation-induced secreted proteins that had higher (up to 12-fold) secretion levels at 24 h post-2 Gy radiation were identified. Secretomic results were combined with whole-transcriptome gene expression experiments, using both radiosensitive and radioresistant cells, to identify a signature related to intrinsic radiosensitivity. Gene expression analysis assessing the levels of the 33 proteins showed that 5 (YBX3, EIF4EBP2, DKK1, GNPNAT1 and TK1) had higher expression levels in the radiosensitive cells compared to their radioresistant derivatives; 3 of these proteins (DKK1, GNPNAT1 and TK1) underwent in-lab and initial clinical validation. Western blot analysis using CM samples from cell lines confirmed a significant increase in the release of each candidate biomarker from radiosensitive cells 24 h after treatment with a 2 Gy dose of radiation; no significant increase in secretion was observed in the radioresistant cells after radiation. Immunohistochemistry showed that higher intracellular protein levels of the biomarkers were associated with greater radiosensitivity. Intracellular levels were further assessed in pre-treatment biopsy tissues from patients diagnosed with ER+ BC that were subsequently treated with breast-conserving surgery and RT. High DKK1 and GNPNAT1 intracellular levels were associated with significantly increased recurrence-free survival times, indicating that these two candidate biomarkers have the potential to predict sensitivity to RT. We suggest that the methods highlighted in this study could be utilised for the identification of biomarkers that may have a potential clinical role in personalising and optimising RT dosing regimens, whilst limiting the administration of RT to patients who are unlikely to benefit.
Collapse
|
7
|
|
8
|
Esposito M, Fang C, Cook KC, Park N, Wei Y, Spadazzi C, Bracha D, Gunaratna RT, Laevsky G, DeCoste CJ, Slabodkin H, Brangwynne CP, Cristea IM, Kang Y. TGF-β-induced DACT1 biomolecular condensates repress Wnt signalling to promote bone metastasis. Nat Cell Biol 2021; 23:257-267. [PMID: 33723425 PMCID: PMC7970447 DOI: 10.1038/s41556-021-00641-w] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/26/2021] [Indexed: 02/07/2023]
Abstract
The complexity of intracellular signalling requires both a diversity of molecular players and the sequestration of activity to unique compartments within the cell. Recent findings on the role of liquid-liquid phase separation provide a distinct mechanism for the spatial segregation of proteins to regulate signalling pathway crosstalk. Here, we discover that DACT1 is induced by TGFβ and forms protein condensates in the cytoplasm to repress Wnt signalling. These condensates do not localize to any known organelles but, rather, exist as phase-separated proteinaceous cytoplasmic bodies. The deletion of intrinsically disordered domains within the DACT1 protein eliminates its ability to both form protein condensates and suppress Wnt signalling. Isolation and mass spectrometry analysis of these particles revealed a complex of protein machinery that sequesters casein kinase 2-a Wnt pathway activator. We further demonstrate that DACT1 condensates are maintained in vivo and that DACT1 is critical to breast and prostate cancer bone metastasis.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Cao Fang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Katelyn C Cook
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Nana Park
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Chiara Spadazzi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Dan Bracha
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Ramesh T Gunaratna
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Gary Laevsky
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - Hannah Slabodkin
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Clifford P Brangwynne
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
- Howard Hughes Medical Institute, Princeton University, Princeton, NJ, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA.
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
9
|
Zhu G, Song J, Chen W, Yuan D, Wang W, Chen X, Liu H, Su H, Zhu J. Expression and Role of Dickkopf-1 (Dkk1) in Tumors: From the Cells to the Patients. Cancer Manag Res 2021; 13:659-675. [PMID: 33536782 PMCID: PMC7847771 DOI: 10.2147/cmar.s275172] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 09/26/2020] [Indexed: 12/14/2022] Open
Abstract
Dickkopf-1 (Dkk1) is a secretory antagonist of the classical Wnt signaling pathway. Many studies have reported that Dkk1 is abnormally expressed in tumor cells, and abnormal expression of Dkk1 can inhibit cell proliferation or induce apoptosis through pro-apoptotic factors, However, due to the differences in tumor environment and the complex regulatory mechanisms in different tumors, Dkk1 has different effects on the progression of different tumors. In many tumors, high expression of Dkk1 may promote tumor metastasis. However, Dkk1, which is highly expressed in other tumors, can inhibit tumor invasion and metastasis. More and more evidence shows that Dkk1 plays a complex and different role in tumor occurrence, development and metastasis in different tumor environments and through a variety of complex regulatory mechanisms. Therefore, Dkk1 may not only be a useful biomarker of metastasis, but also a target for studying the metabolic mechanism of tumor cells and treating tumors in many tumor types. Therefore, this article reviews the research progress on the expression, mechanism and function of Dkk1 in different tumors, and at the same time, based on the public database data, we made a further analysis of the expression of Dkk1 in different tumors.
Collapse
Affiliation(s)
- Guohua Zhu
- Guizhou Medical University, Guiyang, Guizhou Province 550002, People's Republic of China.,Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province 550002, People's Republic of China
| | - Jukun Song
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province 550002, People's Republic of China.,Guizhou University School of Medicine, Guiyang, Guizhou Province 550025, People's Republic of China
| | - Weimin Chen
- Guizhou University School of Medicine, Guiyang, Guizhou Province 550025, People's Republic of China
| | - Dongbo Yuan
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province 550002, People's Republic of China.,Guizhou University School of Medicine, Guiyang, Guizhou Province 550025, People's Republic of China
| | - Wei Wang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province 550002, People's Republic of China
| | - Xiaoyue Chen
- Guizhou University School of Medicine, Guiyang, Guizhou Province 550025, People's Republic of China
| | - Hen Liu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province 550002, People's Republic of China.,Zunyi Medical University, Zunyi, Guizhou Province 563000, People's Republic of China
| | - Hao Su
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province 550002, People's Republic of China.,Zunyi Medical University, Zunyi, Guizhou Province 563000, People's Republic of China
| | - Jianguo Zhu
- Guizhou Medical University, Guiyang, Guizhou Province 550002, People's Republic of China.,Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province 550002, People's Republic of China.,Guizhou University School of Medicine, Guiyang, Guizhou Province 550025, People's Republic of China.,Zunyi Medical University, Zunyi, Guizhou Province 563000, People's Republic of China
| |
Collapse
|
10
|
Breast Cancer and Microcalcifications: An Osteoimmunological Disorder? Int J Mol Sci 2020; 21:ijms21228613. [PMID: 33203195 PMCID: PMC7696282 DOI: 10.3390/ijms21228613] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
The presence of microcalcifications in the breast microenvironment, combined with the growing evidences of the possible presence of osteoblast-like or osteoclast-like cells in the breast, suggest the existence of active processes of calcification in the breast tissue during a woman’s life. Furthermore, much evidence that osteoimmunological disorders, such as osteoarthritis, rheumatoid arthritis, or periodontitis influence the risk of developing breast cancer in women exists and vice versa. Antiresorptive drugs benefits on breast cancer incidence and progression have been reported in the past decades. More recently, biological agents targeting pro-inflammatory cytokines used against rheumatoid arthritis also demonstrated benefits against breast cancer cell lines proliferation, viability, and migratory abilities, both in vitro and in vivo in xenografted mice. Hence, it is tempting to hypothesize that breast carcinogenesis should be considered as a potential osteoimmunological disorder. In this review, we compare microenvironments and molecular characteristics in the most frequent osteoimmunological disorders with major events occurring in a woman’s breast during her lifetime. We also highlight what the use of bone anabolic drugs, antiresorptive, and biological agents targeting pro-inflammatory cytokines against breast cancer can teach us.
Collapse
|
11
|
Li J, Gao Y, Yue W. The Clinical Diagnostic and Prognostic Value of Dickkopf-1 in Cancer. Cancer Manag Res 2020; 12:4253-4260. [PMID: 32606922 PMCID: PMC7292247 DOI: 10.2147/cmar.s254596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/19/2020] [Indexed: 12/25/2022] Open
Abstract
The Wnt signaling pathway extensively participates in diverse processes such as embryonic development, maintenance of homeostasis and tumor pathogenesis. Dickkopf-1 (DKK1), a Wnt inhibitor, plays a vital role for over the past decades regarding its role in the regulation of several types of cancers. However, studies have shown that DKK1 is expressed differently in cancer and plays a role as a cancer-promoting factor or a tumor suppressor, which is worthy of further exploration. We herein study whether DKK1 is highly expressed in all cancers and plays a crucial role in promoting cancer. Furthermore, we discussed as to which stages of cancer development it plays in. Finally, the present detection methods were introduced and indicated the clinical application of DKK1 in tumor development.
Collapse
Affiliation(s)
- Jie Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| | - Yan Gao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| | - Wentao Yue
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| |
Collapse
|
12
|
Song Z, Wang H, Zhang S. Negative regulators of Wnt signaling in non-small cell lung cancer: Theoretical basis and therapeutic potency. Biomed Pharmacother 2019; 118:109336. [PMID: 31545260 DOI: 10.1016/j.biopha.2019.109336] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/20/2019] [Accepted: 08/05/2019] [Indexed: 02/05/2023] Open
Abstract
Significant advances in the treatment of non-small cell lung cancer (NSCLC) have been made over the past decade, and they predominantly involve molecular targets such as epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements. However, despite the initial good response, drug resistance eventually develops. The Wnt signaling pathway has recently been considered important in embryonic development and tumorigenesis in many cancers, particularly NSCLC. Moreover, the aberrant Wnt pathway plays a significant role in NSCLC and is associated with cancer cell proliferation, metastasis, invasion and drug resistance, and the suppression of canonical or noncanonical Wnt signaling through various biological or pharmacological negative regulators has been proven to produce specific anticancer effects. Thus, blocking the Wnt pathway via its negative regulators may overcome the resistance of current treatment methods and lead to new treatment strategies for NSCLC. Therefore, in this review, we summarize recent studies on the role of negative regulators in Wnt signaling in NSCLC and the therapeutic potency of these molecules as agents and targets for NSCLC treatments.
Collapse
Affiliation(s)
- Zikuan Song
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haoyu Wang
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuang Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Zhang J, Li WY, Yang Y, Yan LZ, Zhang SY, He J, Wang JX. LncRNA XIST facilitates cell growth, migration and invasion via modulating H3 histone methylation of DKK1 in neuroblastoma. Cell Cycle 2019; 18:1882-1892. [PMID: 31208278 PMCID: PMC6681787 DOI: 10.1080/15384101.2019.1632134] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been confirmed to be aberrantly expressed and involved in the progression of neuroblastoma. This study aimed to explore the expression profile of lncRNA X-inactive specific transcript (XIST) and its functional involvement in neuroblastoma. In this study, the relative level of XIST in neuroblastoma tissues and cell lines was detected by qPCR, and DKK1 protein expression was determined using western blot. The effect of XIST on cell growth, invasion and migration in vitro and in tumorigenesis of neuroblastoma was assessed. The level of H3K27me3 in DKK1 promoter was analyzed with ChIP-qPCR. Interaction between XIST and EZH2 was verified by RNA immunoprecipitation (RIP) and RNA pull-down assay. XIST was significantly upregulated in neuroblastoma tissues (n = 30) and cells lines, and it was statistically associated with the age and International Neuroblastoma Staging System (INSS) staging in neuroblastoma patients. Downregulation of XIST suppressed the growth, migration and invasion of neuroblastoma cells. EZH2 inhibited DKK1 expression through inducing H3 histone methylation in its promoter. XIST increased the level of H3K27me3 in DKK1 promoter via interacting with EZH2. Downregulation of XIST increased DKK1 expression to suppress neuroblastoma cell growth, invasion, and migration, which markedly restrained the tumor progression. In conclusion, XIST downregulated DKK1 by inducing H3 histone methylation via EZH2, thereby facilitating the growth, migration and invasion of neuroblastoma cells and retarding tumor progression.
Collapse
Affiliation(s)
- Jiao Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- CONTACT Jiao Zhang
| | - Wen-Ya Li
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Yang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li-Zhao Yan
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Song-Yang Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jia-Xiang Wang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
14
|
Schisandrin A inhibits triple negative breast cancer cells by regulating Wnt/ER stress signaling pathway. Biomed Pharmacother 2019; 115:108922. [DOI: 10.1016/j.biopha.2019.108922] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/24/2019] [Accepted: 04/24/2019] [Indexed: 01/09/2023] Open
|
15
|
Zhuang X, Zhang H, Hu G. Cancer and Microenvironment Plasticity: Double-Edged Swords in Metastasis. Trends Pharmacol Sci 2019; 40:419-429. [PMID: 31078320 DOI: 10.1016/j.tips.2019.04.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/07/2019] [Accepted: 04/12/2019] [Indexed: 02/07/2023]
Abstract
Cancer initiates at one site (primary tumor) and, in most cases, spreads to other distant organs (metastasis). During the multistep process of metastasis, primary tumor cells acquire cellular and phenotypic plasticity to survive and thrive in different environments. Moreover, cancer cells also utilize and educate microenvironmental components by reshaping them into accomplices of metastasis. Recent studies have identified a plethora of new molecular and cellular modulators of metastasis that have dynamic or even opposite roles, dominating the phenotypic plasticity of both tumoral and microenvironmental components. In this review we discuss their bipotential functions and the possible underpinning mechanisms, as well as their implications for targeted cancer therapy.
Collapse
Affiliation(s)
- Xueqian Zhuang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hao Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
16
|
Cucchi D, Menon A, Galliera E, Messina C, Zanini B, Marazzi MG, Massaccesi L, Compagnoni R, Corsi Romanelli MM, Randelli P. A Prospective Assessment of Periprosthetic Bone Mineral Density and Osteoimmunological Biomarkers Variations After Total Knee Replacement Surgery. J Clin Densitom 2019; 22:86-95. [PMID: 30072203 DOI: 10.1016/j.jocd.2018.05.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
Aseptic loosening is a major cause of premature failure of total knee replacement (TKR). Variations in periprosthetic bone mineral density (BMD) and osteoimmunological biomarkers levels could help to quantify prosthesis osteointegration and predict early aseptic loosening. The gene expression of 5 selected osteoimmunological biomarkers was evaluated in tibial plateau bone biopsies by real-time polymerase chain reaction and changes in their serum levels after TKR were prospectively evaluated with enzyme-linked immunosorbent assay for 1 yr after surgery. These variations were correlated to changes in periprosthetic BMD. Sixteen patients were evaluated. A statistically significant decrease in serum levels of Sclerostin (p = 0.0135) was observed immediately after surgery. A specular pattern was observed between dickkopf-related protein 1 and osteoprotegerin expression. No statistically significant changes were detectable in the other study biomarkers. Periprosthetic BMD did not change significantly across the duration of the follow-up. Prosthetic knee surgery has an impact on bone remodeling, in particular on sclerostin expression. Although not showing statistically significant changes, in the patterns of dickkopf-related protein 1, osteoprotegerin, and the ligand of the receptor activator of nuclear factor kappa-B symmetries and correspondences related to the biological activities of these proteins could be identified. Variation in osteoimmunological biomarkers after TKR surgery can help in quantifying prosthesis osteointegration.
Collapse
Affiliation(s)
- Davide Cucchi
- Department of Orthopaedics and Trauma Surgery, Universitätsklinikum Bonn, Bonn 53127, Germany; Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan 20133, Italy.
| | - Alessandra Menon
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan 20133, Italy; 1° Clinica Ortopedica, ASST Centro Specialistico Ortopedico Traumatologico Gaetano Pini-CTO, Milan 20122, Italy
| | - Emanuela Galliera
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan 20133, Italy; IRCCS Galeazzi Orthopaedic Institute, Milan 20161, Italy
| | - Carmelo Messina
- Department of Diagnostic and Interventional Radiology, IRCCS Galeazzi Orthopaedic Institute, Milan 20161, Italy
| | - Beatrice Zanini
- 1° Clinica Ortopedica, ASST Centro Specialistico Ortopedico Traumatologico Gaetano Pini-CTO, Milan 20122, Italy
| | - Monica Gioia Marazzi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan 20133, Italy
| | - Luca Massaccesi
- Department of Biomedical, Surgical and Oral Science, Università degli Studi di Milano, Milan 20133, Italy
| | - Riccardo Compagnoni
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan 20133, Italy; 1° Clinica Ortopedica, ASST Centro Specialistico Ortopedico Traumatologico Gaetano Pini-CTO, Milan 20122, Italy
| | - Massimiliano M Corsi Romanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan 20133, Italy; U.O.C SMEL-1 Patologia Clinica, IRCCS Policlinico San Donato, Milan 20097, Italy
| | - Pietro Randelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan 20133, Italy; 1° Clinica Ortopedica, ASST Centro Specialistico Ortopedico Traumatologico Gaetano Pini-CTO, Milan 20122, Italy
| |
Collapse
|
17
|
Galliera E, Ragone V, Marazzi MG, Selmin F, Banci L, Corsi Romanelli MM. Vitamin E-stabilized UHMWPE: Biological response on human osteoblasts to wear debris. Clin Chim Acta 2018; 486:18-25. [DOI: 10.1016/j.cca.2018.07.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/09/2018] [Indexed: 12/31/2022]
|
18
|
Dickkopf-1 (Dkk1) protein expression in breast cancer with special reference to bone metastases. Clin Exp Metastasis 2018; 35:763-775. [PMID: 30238177 DOI: 10.1007/s10585-018-9937-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/18/2018] [Indexed: 12/15/2022]
Abstract
Dysregulation of the Wnt inhibitor dickkopf-1 protein (Dkk1) has been reported in a variety of cancers. In addition, it has been linked to the progression of malignant bone disease by impairing osteoblast activity. This study investigated serum- and tissue levels of Dkk1 in breast cancer patients with- or without bone metastases. Serum Dkk1 levels were measured by ELISA in 89 breast cancer patients and 86 healthy women. Tissue levels of Dkk1 and β-catenin, a major downstream component of Wnt transduction pathway, were tested with immunohistochemical staining in 143 different tissues, including adjacent non-tumoral breast tissues, primary breast tumours, lymph nodes metastases, and bone metastases. Serum levels of Dkk1 were significantly increased in breast cancer patients without metastases compared with healthy controls and even more increased in patients with bone metastases. Tissue expression of Dkk1 was positive in 70% of tested primary breast cancer tissues and demonstrated significant correlation with histological type and PR status. Less frequent expression of Dkk1 was found in lymph nodes metastases and bone metastases compared with adjacent non-tumoral breast tissues and primary breast tumours. Tissue expression of β-catenin was positive in the vast majority of all tested tissue types indicating activated Wnt/β-catenin signalling. Our results suggested that Wnt/β-catenin signalling in breast tumours and their secondary lymph nodes- and bone metastases is dysregulated and this could be related to aberrant Dkk1 expression levels. Hence, Dkk1 protein might provide insights into the continued development of novel comprehensive and therapeutic strategies for breast cancer and its bone metastases.
Collapse
|
19
|
Li X, Yang J, Bao M, Zeng K, Fu S, Wang C, Ye L. Wnt signaling in bone metastasis: mechanisms and therapeutic opportunities. Life Sci 2018; 208:33-45. [PMID: 29969609 DOI: 10.1016/j.lfs.2018.06.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/29/2018] [Accepted: 06/29/2018] [Indexed: 02/05/2023]
Abstract
Bone metastasis frequently occurs in advanced cancer patients, who will develop osteogenic/osteolytic bone lesions in the late stage of the disease. Wnt signaling pathway, which is mainly grouped into the β-catenin dependent pathway and β-catenin independent pathway, is a well-organized cascade that has been reported to play important roles in a variety of physiological and pathological conditions, including bone metastasis. Regulation of Wnt signaling in bone metastasis involves multiple stages, including dissemination of primary tumor cells to bone, dormancy and outgrowth of metastatic tumor cells, and tumor-induced osteogenic and osteolytic bone destruction, suggesting the importance of Wnt signaling in bone metastasis pathology. In this review, we will introduce the involvement of Wnt signaling components in specific bone metastasis stages and summarize the promising Wnt modulators that have shown potential as bone metastasis therapeutics, in the hope to maximize the therapeutic opportunities of Wnt signaling for bone metastasis.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Minyue Bao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kan Zeng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shijin Fu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenglin Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
20
|
Kreutz D, Sinthuvanich C, Bileck A, Janker L, Muqaku B, Slany A, Gerner C. Curcumin exerts its antitumor effects in a context dependent fashion. J Proteomics 2018; 182:65-72. [PMID: 29751106 DOI: 10.1016/j.jprot.2018.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/29/2018] [Accepted: 05/04/2018] [Indexed: 02/08/2023]
Abstract
Proteome profiling profoundly contributes to the understanding of cell response mechanisms to drug actions. Such knowledge may become a key to improve personalized medicine. In the present study, the effects of the natural remedy curcumin on breast cancer model systems were investigated. MCF-7, ZR-75-1 and TGF-β1 pretreated fibroblasts, mimicking cancer-associated fibroblasts (CAFs), were treated independently as well as in tumor cell/CAF co-cultures. Remarkably, co-culturing with CAF-like cells (CLCs) induced different proteome alterations in MCF-7 and ZR-75-1 cells, respectively. Curcumin significantly induced HMOX1 in single cell type models and co-cultures. However, other curcumin effects differed. In the MCF-7/CLC co-culture, curcumin significantly down-regulated RC3H1, a repressor of inflammatory signaling. In the ZR-75-1/CLC co-culture, curcumin significantly down-regulated PEG10, an anti-apoptotic protein, and induced RRAGA, a pro-apoptotic protein involved in TNF-alpha signaling. Furthermore, curcumin induced AKR1C2, an important enzyme for progesterone metabolism. None of these specific curcumin effects were observed in single cell type cultures. All high-resolution mass spectrometry data are available via ProteomeXchange with the identifier PXD008719. The present data demonstrate that curcumin induces proteome alterations, potentially accounting for its known antitumor effects, in a strongly context-dependent fashion. BIOLOGICAL SIGNIFICANCE Better means to understand and potentially predict individual variations of drug effects are urgently required. The present proteome profiling study of curcumin effects demonstrates the massive impact of the cell microenvironment on cell responses to drug action. Co-culture models apparently provide more biologically relevant information regarding curcumin effects than single cell type cultures.
Collapse
Affiliation(s)
- Dominique Kreutz
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Chomdao Sinthuvanich
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Besnik Muqaku
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Astrid Slany
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria.
| |
Collapse
|
21
|
Chean J, Chen CJ, Shively JE. ETS transcription factor ELF5 induces lumen formation in a 3D model of mammary morphogenesis and its expression is inhibited by Jak2 inhibitor TG101348. Exp Cell Res 2017; 359:62-75. [PMID: 28800960 DOI: 10.1016/j.yexcr.2017.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 08/02/2017] [Accepted: 08/05/2017] [Indexed: 02/06/2023]
Abstract
The loss of expression of a single gene can revert normal tissue to a malignant phenotype. For example, while normal breast has high lumenal expression of CEACAM1, the majority of breast cancers exhibit the early loss of this gene with the concurrent loss of their lumenal phenotype. MCF7 cells that lack CEACAM1 expression and fail to form lumena in 3D culture, regain the normal phenotype when transfected with CEACAM1. In order to probe the mechanism of this gain of function, we treated these cells with the clinically relevant Jak2 inhibitor TG101348 (TG), expecting that disruption of the prolactin receptor signaling pathway would interfere with the positive effects of transfection of MCF7 cells with CEACAM1. Indeed, lumen formation was inhibited, resulting in the down regulation of a set of genes, likely involved in the complex process of lumen formation. As expected, inhibition of the expression of many of these genes also inhibited lumen formation, confirming their involvement in a single pathway. Among the genes identified by the inhibition assay, ETS transcription factor ELF5 stood out, since it has been identified as a master regulator of mammary morphogenesis, and is associated with prolactin receptor signaling. When ELF5 was transfected into the parental MCF7 cells that lack CEACAM1, lumen formation was restored, indicating that ELF5 can replace CEACAM1 in this model system of lumenogenesis. We conclude that the event(s) that led to the loss of expression of CEACAM1 is epistatic in that multiple genes associated with a critical pathway were affected, but that restoration of the normal phenotype can be achieved with reactivation of certain genes at various nodal points in tissue morphogenesis.
Collapse
Affiliation(s)
- Jennifer Chean
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, 1450 E. Duarte Road, Duarte, CA 91010, USA
| | - Charng-Jui Chen
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, 1450 E. Duarte Road, Duarte, CA 91010, USA
| | - John E Shively
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, 1450 E. Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
22
|
Differential effects on lung and bone metastasis of breast cancer by Wnt signalling inhibitor DKK1. Nat Cell Biol 2017; 19:1274-1285. [PMID: 28892080 DOI: 10.1038/ncb3613] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 08/14/2017] [Indexed: 12/14/2022]
Abstract
Metastatic cancer is a systemic disease, and metastasis determinants might elicit completely different effects in various target organs. Here we show that tumour-secreted DKK1 is a serological marker of breast cancer metastasis organotropism and inhibits lung metastasis. DKK1 suppresses PTGS2-induced macrophage and neutrophil recruitment in lung metastases by antagonizing cancer cell non-canonical WNT/PCP-RAC1-JNK signalling. In the lungs, DKK1 also inhibits WNT/Ca2+-CaMKII-NF-κB signalling and suppresses LTBP1-mediated TGF-β secretion of cancer cells. In contrast, DKK1 promotes breast-to-bone metastasis by regulating canonical WNT signalling of osteoblasts. Importantly, targeting canonical WNT may not be beneficial to treatment of metastatic cancer, while combinatory therapy against JNK and TGF-β signalling effectively prevents metastasis to both the lungs and bone. Thus, DKK1 represents a class of Janus-faced molecules with dichotomous roles in organotropic metastasis, and our data provide a rationale for new anti-metastasis approaches.
Collapse
|
23
|
The role of miR-372 in ovarian carcinoma cell proliferation. Gene 2017; 624:14-20. [DOI: 10.1016/j.gene.2017.04.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 04/25/2017] [Indexed: 01/22/2023]
|
24
|
Shen J, Chen X, Jia H, Meyers CA, Shrestha S, Asatrian G, Ding C, Tsuei R, Zhang X, Peault B, Ting K, Soo C, James AW. Effects of WNT3A and WNT16 on the Osteogenic and Adipogenic Differentiation of Perivascular Stem/Stromal Cells. Tissue Eng Part A 2017; 24:68-80. [PMID: 28463594 DOI: 10.1089/ten.tea.2016.0387] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Human perivascular stem/stromal cells (hPSC) are a multipotent mesenchymogenic stromal cell population defined by their perivascular locale. Recent studies have demonstrated the high potential for clinical translation of this fluorescence-activated cell sorting (FACS)-derived cell population for autologous bone tissue engineering. However, the mechanisms underlying the osteogenic differentiation of PSC are incompletely understood. The current study investigates the roles of canonical and noncanonical Wnt signaling in the osteogenic and adipogenic differentiation of PSC. Results showed that both canonical and noncanonical Wnt signaling activity transiently increased during PSC osteogenic differentiation in vitro. Sustained WNT3A treatment significantly decreased PSC osteogenic differentiation. Conversely, sustained treatment with Wnt family member 16 (WNT16), a mixed canonical and noncanonical ligand, increased osteogenic differentiation in a c-Jun N-terminal kinase (JNK) pathway-dependent manner. Conversely, WNT16 knockdown significantly diminished PSC osteogenic differentiation. Finally, WNT16 but not WNT3A increased the adipogenic differentiation of PSC. These results indicate the importance of regulation of canonical and noncanonical Wnt signaling for PSC fate and differentiation. Moreover, these data suggest that WNT16 plays a functional and necessary role in PSC osteogenesis.
Collapse
Affiliation(s)
- Jia Shen
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Xuepeng Chen
- 2 Department of Orthodontics, Stomatological Hospital, Zhejiang University , Hangzhou, China
| | - Haichao Jia
- 3 Department of Orthodontics, School of Stomatology, Capital Medical University , Beijing, China
| | - Carolyn A Meyers
- 4 Department of Pathology, Johns Hopkins University , Baltimore, Maryland
| | - Swati Shrestha
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Greg Asatrian
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Catherine Ding
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Rebecca Tsuei
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Xinli Zhang
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Bruno Peault
- 5 Department of Orthopaedic Surgery, UCLA and Orthopaedic Hospital, Orthopaedic Hospital Research Center , Los Angeles, California.,6 Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh , Edinburgh, United Kingdom
| | - Kang Ting
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA , Los Angeles, California
| | - Chia Soo
- 5 Department of Orthopaedic Surgery, UCLA and Orthopaedic Hospital, Orthopaedic Hospital Research Center , Los Angeles, California.,7 Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California , Los Angeles, Los Angeles, California
| | - Aaron W James
- 4 Department of Pathology, Johns Hopkins University , Baltimore, Maryland.,5 Department of Orthopaedic Surgery, UCLA and Orthopaedic Hospital, Orthopaedic Hospital Research Center , Los Angeles, California
| |
Collapse
|
25
|
|
26
|
Pang H, Ma N, Jiao M, Shen W, Xin B, Wang T, Zhang F, Liu L, Zhang H. The Biological Effects of Dickkopf1 on Small Cell Lung Cancer Cells and Bone Metastasis. Oncol Res 2017; 25:35-42. [PMID: 28081731 PMCID: PMC7840681 DOI: 10.3727/096504016x14719078133249] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The bone is among the most common sites of metastasis in patients with lung cancer. Over 30%–40% of lung cancers can develop bone metastasis, and no effective therapeutic methods exist in clinic cases. Wnt/β-catenin signaling and Dickkopf1 (DKK1) play important roles in the progression of lung cancer, which preferentially metastasizes to the skeleton. However, the role of DKK1 in osteotropism of small cell lung cancer (SCLC) remains to be elucidated. This study aimed to define the role of DKK1 in SCLC bone metastasis and investigate the underlying mechanisms. Our results demonstrated that the expression level of DKK1 was dramatically higher in bone metastatic SCLC cells (SBC-5 cell line) compared with that in cells without bone metastatic ability (SBC-3 cell line). Therefore, we hypothesized that DKK1 was involved in the bone metastasis of SCLC. We then suppressed the DKK1 expression in SBC-5 cells by RNAi and found that downregulation of DKK1 can inhibit cell proliferation, colony formation, cell migration, and invasion, but increase the apoptosis rate. Downregulation of DKK1 did not affect the cell cycle progression of SBC-5 cells in vitro. In vivo, downregulated DKK1 in SBC-5 cells resulted in attenuated bone metastasis. These results indicated that DKK1 may be an important regulator in bone metastases of SCLC, and targeting DKK1 may be an effective method to prevent and treat skeleton metastases in SCLC cases.
Collapse
|
27
|
EGb-761 Attenuates the Anti-proliferative Activity of Fluoride via DDK1 in PC-12 Cells. Neurochem Res 2016; 42:606-614. [DOI: 10.1007/s11064-016-2115-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/11/2016] [Accepted: 11/16/2016] [Indexed: 01/08/2023]
|
28
|
Göbel A, Thiele S, Browne AJ, Rauner M, Zinna VM, Hofbauer LC, Rachner TD. Combined inhibition of the mevalonate pathway with statins and zoledronic acid potentiates their anti-tumor effects in human breast cancer cells. Cancer Lett 2016; 375:162-171. [DOI: 10.1016/j.canlet.2016.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 12/22/2022]
|
29
|
Li X, Xiao R, Tembo K, Hao L, Xiong M, Pan S, Yang X, Yuan W, Xiong J, Zhang Q. PEG10 promotes human breast cancer cell proliferation, migration and invasion. Int J Oncol 2016; 48:1933-42. [PMID: 26934961 DOI: 10.3892/ijo.2016.3406] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 01/08/2016] [Indexed: 11/05/2022] Open
Abstract
Paternally expressed imprinted gene 10 (PEG10), derived from the Ty3/Gypsy family of retrotransposons, has been implicated as a genetic imprinted gene. Accumulating evidence suggests that PEG10 plays an important role in tumor growth in various cancers, including hepatocellular carcinoma, lung cancer and prostate cancer. However, the correlation between PEG10 and breast cancer remains unclear. In the present study, we evaluated and characterized the role of PEG10 in human breast cancer proliferation, cell cycle, clone formation, migration and invasion. The expression level of PEG10 was significantly elevated in breast cancer tissues and associated with distant metastasis and poor clinical outcome. Gene set enrichment analysis indicated that high expression of PEG10 could enrich cell cycle-related processes in breast cancer tissues. Ectopic overexpression of PEG10 in breast cancer cells enhanced cell proliferation, cell cycle, clone formation along with migration and invasion. Cell-to-cell junction molecule E-cadherin was downregulated and matrix degradation proteases MMP-1, MMP-2, MMP-9 were up-regulated after PEG10 overexpression. Our results demonstrated that PEG10 is a crucial oncogene and has prognostic value for breast cancer, which could be applied in breast cancer diagnosis and targeting therapy in future.
Collapse
Affiliation(s)
- Xinran Li
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ruijing Xiao
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Kingsley Tembo
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ling Hao
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Meng Xiong
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Shan Pan
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xiangyong Yang
- Hubei University of Technology Engineering and Technology College, Wuhan, Hubei 430068, P.R. China
| | - Wen Yuan
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jie Xiong
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Qiuping Zhang
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|