1
|
Ma Y, Gong H, Cheng L, Zhang D. Discoid Domain Receptors Signaling in Macrophages-Mediated Diseases. Int J Gen Med 2025; 18:907-926. [PMID: 39990299 PMCID: PMC11847422 DOI: 10.2147/ijgm.s487093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/18/2025] [Indexed: 02/25/2025] Open
Abstract
Macrophages, as a crucial component of the body's immune system, play a vital role in the onset, progression, and outcome of diseases. Discoidin domain receptors (DDRs), important members of the novel receptor tyrosine kinase superfamily, exhibit unique functions in macrophage physiology. Through interactions with the extracellular matrix, DDRs activate signaling pathways such as p38 MAPK and NF-κB, regulating macrophage adhesion, migration, and secretory functions, thereby influencing their behavior in diseases. Recent studies have indicated a direct correlation between DDRs and the progression of various diseases, including inflammation, cancer, and fibrosis. However, there remain numerous knowledge gaps regarding the specific mechanisms by which DDRs function in macrophage-mediated diseases. This article provides an in-depth summary of the regulatory mechanisms of DDRs on macrophages, detailing their modulatory roles in various diseases through macrophages and their underlying mechanisms. The aim is to offer new insights into biomedical therapies targeting DDRs and the development of novel drugs.
Collapse
Affiliation(s)
- Yaohui Ma
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Hang Gong
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Long Cheng
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Dekui Zhang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| |
Collapse
|
2
|
Sengupta S, Maji L, Das PK, Teli G, Nag M, Khan N, Haque M, Matada GSP. Explanatory review on DDR inhibitors: their biological activity, synthetic route, and structure-activity relationship. Mol Divers 2025:10.1007/s11030-024-11091-5. [PMID: 39883387 DOI: 10.1007/s11030-024-11091-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
Discoidin domain receptors (DDR) are categorized under tyrosine kinase receptors (RTKs) and play a crucial role in various etiological conditions such as cancer, fibrosis, atherosclerosis, osteoarthritis, and inflammatory diseases. The structural domain rearrangement of DDR1 and DDR2 involved six domains of interest namely N-terminal DS, DS-like, intracellular juxtamembrane, transmembrane juxtamembrane, extracellular juxtamembrane intracellular kinase domain, and the tail portion contains small C-tail linkage. DDR has not been explored to a wide extent to be declared as a prime target for any particular pathological condition. Very few scientific data are available so there is a need to study the receptors and their inhibitors. Still, there did not exist FDA-approved small molecules targeting DDR1 and DDR2 receptors so there is an urgent need to develop potent small molecules. Further, the structural features and ligand specificities encourage the researchers to be fascinated about the DDR and explore them for the mentioned biological conditions. Therefore, in the last few years, researchers have been involved in investigating the potent DDR inhibitors. The current review provides an outlook on the anatomy and physiology of DDR, focusing on the structural features of DDR receptors and the mechanism of signaling pathways. We have also compiled the evolutionary development status of DDR inhibitors according to their chemical classes, biological activity, selectivity, and structure-activity relationship. From biological activity analysis, it was revealed that compounds 64a (selectivity: DDR1) and 103a (selectivity: DDR2) were the most potent candidates with excellent activity with IC50 values of 4.67 and 3.2 nM, respectively.
Collapse
Affiliation(s)
- Sindhuja Sengupta
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
- NEF College of Pharmaceutical Education & Research, Nagaon, 782001, India
| | - Lalmohan Maji
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
- Tarifa Memorial Institute of Pharmacy, Murshidabad, West Bengal, 742166, India
| | - Pronoy Kanti Das
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Ghanshyam Teli
- School of Pharmacy, Sangam University, Atoon, Bhilwara, Rajasthan, 311001, India.
| | - Mrinmoy Nag
- NEF College of Pharmaceutical Education & Research, Nagaon, 782001, India
| | - Nirmalya Khan
- Tarifa Memorial Institute of Pharmacy, Murshidabad, West Bengal, 742166, India
| | - Mridul Haque
- Tarifa Memorial Institute of Pharmacy, Murshidabad, West Bengal, 742166, India
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| |
Collapse
|
3
|
Soloveva N, Novikova S, Farafonova T, Tikhonova O, Zgoda V. Secretome and Proteome of Extracellular Vesicles Provide Protein Markers of Lung and Colorectal Cancer. Int J Mol Sci 2025; 26:1016. [PMID: 39940785 PMCID: PMC11816676 DOI: 10.3390/ijms26031016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Colorectal cancer (CRC) and lung cancer (LC) are leading causes of cancer-related mortality, highlighting the need for minimally invasive diagnostic, prognostic, and predictive markers for these cancers. Proteins secreted by a tumor into the extracellular space directly, known as the tumor secretome, as well as proteins in the extra-cellular vesicles (EVs), represent an attractive source of biomarkers for CRC and LC. We performed proteomic analyses on secretome and EV samples from LC (A549, NCI-H23, NCI-H460) and CRC (Caco2, HCT116, HT-29) cell lines and targeted mass spectrometry on EVs from plasma samples of 20 patients with CRC and 19 healthy controls. A total of 782 proteins were identified across the CRC and LC secretome and EV samples. Of these, 22 and 44 protein markers were significantly elevated in the CRC and LC samples, respectively. Functional annotation revealed enrichment in proteins linked to metastasis and tumor progression for both cancer types. In EVs isolated from the plasma of patients with CRC, ITGB3, HSPA8, TUBA4A, and TLN1 were reduced, whereas FN1, SERPINA1, and CST3 were elevated, compared to healthy controls. These findings support the development of minimally invasive liquid biopsy methods for the detection, prognosis, and treatment monitoring of LC and CRC.
Collapse
Affiliation(s)
| | | | | | | | - Victor Zgoda
- Laboratory of Systems Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (N.S.); (S.N.); (T.F.); (O.T.)
| |
Collapse
|
4
|
Damavandi Z, Riahi P, Majidizadeh T, Houshmand M. Evaluation of t-DARPP Expression Alteration in Association with DDR1 Expression in Non-Small Cell Lung Cancer. IRANIAN BIOMEDICAL JOURNAL 2024; 28:23-30. [PMID: 38308500 PMCID: PMC10994641 DOI: 10.61186/ibj.3878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 06/21/2023] [Indexed: 02/04/2024]
Abstract
Background Discoidin domain receptor 1 (DDR1) signaling plays a critical role in various cellular functions. Increased DDR1 expression has been shown in different human cancers. t-DARPP is a truncated isoform of DARPP-32, and its upregulation promotes cell survival and migration. Most lung cancer patients have non-small cell lung cancer (NSCLC), and their survival rate is low. Therefore, it is necessary to study new and effective targeted therapies. Increased t-DARPP expression in NSCLC patients is associated with patient survival and can act as a prognostic marker correlated with increasing stages of NSCLC. The current study aimed to evaluate alteration in DDR1 expression and its effects on t-DARPP expression in NSCLC. Methods Two human lung adenocarcinoma cell lines, A549 and Calu-3, were treated with collagen type I and transfected with DDR1 siRNA. The relative expression of DDR1 and t-DARPP was evaluated using qRT-PCR. Results The results indicated that collagen type I could stimulate DDR1 expression in NSCLC cells. Also, DDR1 upregulation resulted in a significant increase in t-DARPP expression. In contrast, suppression of DDR1 expression significantly decreased t-DARPP expression. Conclusion Our findings propose that modification in the expression of DDR1, caused by collagen type I and siRNA, might influence the expression of t-DARPP in NSCLC that is linked to NSCLC progression. Moreover, this alteration could potentially serve as an innovative target for therapeutic intervention.
Collapse
Affiliation(s)
| | | | | | - Massoud Houshmand
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
5
|
Vigoda M, Mathieson C, Evans N, Hale C, Jennings J, Lucero O, Jeng S, Bottomly D, Clayburgh D, Andersen P, Li R, Petrisor D, Tyner JW, McWeeney S, Kulesz-Martin M. Functional proteomics of patient derived head and neck squamous cell carcinoma cells reveal novel applications of trametinib. Cancer Biol Ther 2022; 23:310-318. [PMID: 35343367 PMCID: PMC8966983 DOI: 10.1080/15384047.2022.2055420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In this study, we report a differential response of mitogen-activated protein kinase–kinase (MEK) inhibitor trametinib in 20 head and neck squamous cell carcinoma (HNSCC) patients’ tumor-derived cell cultures. Relatively sensitive and resistant cases to trametinib were identified using high throughput metabolic assays and validated in extended dose response studies in vitro. High throughput metabolic assays exploring combination therapies with trametinib were subjected to synergy models and maximal synergistic dose analyses. These yielded several candidates, including axtinib, GDC-0032, GSK-690693, and SGX-523. The combination regimen of trametinib and AXL/MET/VEGFR inhibitor glesatinib showed initial efficacy both in vitro and in vivo (92% reduction in tumor volume). Sensitivity was validated in vivo in a patient-derived xenograft (PDX) model in which trametinib as a single agent effected reduction in tumor volume up to 72%. Reverse Phase Protein Arrays (RPPA) demonstrated differentially expressed proteins and phosphoproteins upon trametinib treatment. Furthermore, resistant cell lines showed a compensatory mechanism via increases in MAPK and non-MAPK pathway proteins that may represent targets for future combination regimens. Intrinsic-targeted options have potential to address paucity of medical treatment options for HNSCC cancer patients, enhance response to extrinsic targeted agents, and/or reduce morbidity as neoadjuvant to surgical treatments.
Collapse
Affiliation(s)
- Myles Vigoda
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.,Michigan State University College of Osteopathic Medicine, East Lansing, MI, USA
| | - Chase Mathieson
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - Nathaniel Evans
- Division of Bioinformatics & Computational Biology, Department of Medical Informatics and Clinical Epidemiolog, Oregon Health & Science University, Portland, OR, USA
| | - Carolyn Hale
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - Jennifer Jennings
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - Olivia Lucero
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Sophia Jeng
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR, USA
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Daniel Clayburgh
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Operative Care Division, Portland VA Health Care System, Portland, OR, USA
| | - Peter Andersen
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Operative Care Division, Portland VA Health Care System, Portland, OR, USA
| | - Ryan Li
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Operative Care Division, Portland VA Health Care System, Portland, OR, USA
| | - Daniel Petrisor
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Operative Care Division, Portland VA Health Care System, Portland, OR, USA
| | - Jeffrey W Tyner
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR, USA
| | - Shannon McWeeney
- Division of Bioinformatics & Computational Biology, Department of Medical Informatics and Clinical Epidemiolog, Oregon Health & Science University, Portland, OR, USA
| | - Molly Kulesz-Martin
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
6
|
Karcini A, Lazar IM. The SKBR3 cell-membrane proteome reveals telltales of aberrant cancer cell proliferation and targets for precision medicine applications. Sci Rep 2022; 12:10847. [PMID: 35760832 PMCID: PMC9237123 DOI: 10.1038/s41598-022-14418-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/07/2022] [Indexed: 12/14/2022] Open
Abstract
The plasma membrane proteome resides at the interface between the extra- and intra-cellular environment and through its various roles in signal transduction, immune recognition, nutrient transport, and cell-cell/cell-matrix interactions plays an absolutely critical role in determining the fate of a cell. Our work was aimed at exploring the cell-membrane proteome of a HER2+ breast-cancer cell line (SKBR3) to identify triggers responsible for uncontrolled cell proliferation and intrinsic resources that enable detection and therapeutic interventions. To mimic environmental conditions that enable cancer cells to evolve adaptation/survival traits, cell culture was performed under serum-rich and serum-deprived conditions. Proteomic analysis enabled the identification of ~ 2000 cell-membrane proteins. Classification into proteins with receptor/enzymatic activity, CD antigens, transporters, and cell adhesion/junction proteins uncovered overlapping roles in processes that drive cell growth, apoptosis, differentiation, immune response, adhesion and migration, as well as alternate pathways for proliferation. The large number of tumor markers (> 50) and putative drug targets (> 100) exposed a vast potential for yet unexplored detection and targeting opportunities, whereas the presence of 15 antigen immunological markers enabled an assessment of epithelial, mesenchymal or stemness characteristics. Serum-starved cells displayed altered processes related to mitochondrial OXPHOS/ATP synthesis, protein folding and localization, while serum-treated cells exhibited attributes that support tissue invasion and metastasis. Altogether, our findings advance the understanding of the biological triggers that sustain aberrant cancer cell proliferation, survival and development of resistance to therapeutic drugs, and reveal vast innate opportunities for guiding immunological profiling and precision medicine applications aimed at target selection or drug discovery.
Collapse
Affiliation(s)
- Arba Karcini
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Iulia M Lazar
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA.
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA.
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA.
| |
Collapse
|
7
|
Wu L, Zhao X, Ma H, Zhang L, Li X. Discoidin Domain Receptor 1, a Potential Biomarker and Therapeutic Target in Hepatocellular Carcinoma. Int J Gen Med 2022; 15:2037-2044. [PMID: 35237068 PMCID: PMC8882470 DOI: 10.2147/ijgm.s348110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 02/10/2022] [Indexed: 11/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is still one of the most lethal human cancers in the world due to its high degree of malignancy, easy invasion and metastasis, poor therapeutic effect and poor prognosis. Nowadays, there is no very effective diagnosis and treatment method. It is crucial to elucidate the underlying pathogenesis and mechanisms of HCC for developing new and effective diagnostic/prognostic biomarkers and therapies. Discoidin domain receptors (DDRs) belong to the family of transmembrane receptor tyrosine kinases (RTKs) and are recognized as playing central regulatory roles in a variety of high incidence human diseases, including tumors. DDRs have two members, DDR1 and DDR2. The role of DDR1 in several tumors has been extensively studied, and many researchers have identified it as a powerful candidate target for the development of functional and effective tumor treatment inhibitors. However, its role and mechanism in HCC are ill defined. In this article, we review the advanced insights into the progression of DDR1 in HCC, particularly the ligands and mechanisms in invasion and metastasis, which may open new avenues for the therapeutic utility of HCC.
Collapse
Affiliation(s)
- Linghong Wu
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Xinhua Zhao
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Huan Ma
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Lili Zhang
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Xiaoan Li
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
- Correspondence: Xiaoan Li, Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 12 Changjia Lane, Jingzhong Street, Fucheng District, Mianyang, 621000, Sichuan, People’s Republic of China, Tel +86 816 224 3593 Email
| |
Collapse
|
8
|
Popescu VB, Kanhaiya K, Năstac DI, Czeizler E, Petre I. Network controllability solutions for computational drug repurposing using genetic algorithms. Sci Rep 2022; 12:1437. [PMID: 35082323 PMCID: PMC8791995 DOI: 10.1038/s41598-022-05335-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/29/2021] [Indexed: 12/22/2022] Open
Abstract
Control theory has seen recently impactful applications in network science, especially in connections with applications in network medicine. A key topic of research is that of finding minimal external interventions that offer control over the dynamics of a given network, a problem known as network controllability. We propose in this article a new solution for this problem based on genetic algorithms. We tailor our solution for applications in computational drug repurposing, seeking to maximize its use of FDA-approved drug targets in a given disease-specific protein-protein interaction network. We demonstrate our algorithm on several cancer networks and on several random networks with their edges distributed according to the Erdős-Rényi, the Scale-Free, and the Small World properties. Overall, we show that our new algorithm is more efficient in identifying relevant drug targets in a disease network, advancing the computational solutions needed for new therapeutic and drug repurposing approaches.
Collapse
Affiliation(s)
| | | | - Dumitru Iulian Năstac
- POLITEHNICA University of Bucharest, Faculty of Electronics, Telecommunications and Information Technology, 061071, Bucharest, Romania
| | - Eugen Czeizler
- Computer Science, Åbo Akademi University, 20500, Turku, Finland
- National Institute for Research and Development in Biological Sciences, 060031, Bucharest, Romania
| | - Ion Petre
- Department of Mathematics and Statistics, University of Turku, 20014, Turku, Finland.
- National Institute for Research and Development in Biological Sciences, 060031, Bucharest, Romania.
| |
Collapse
|
9
|
Curcumin derivative ST09 modulates the miR-199a-5p/DDR1 axis and regulates proliferation and migration in ovarian cancer cells. Sci Rep 2021; 11:23025. [PMID: 34837026 PMCID: PMC8626492 DOI: 10.1038/s41598-021-02454-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/10/2021] [Indexed: 01/04/2023] Open
Abstract
Ovarian cancers are among the fatal malignancies affecting women globally, mainly due to their metastatic and chemoresistant nature. In this study, we report a potent curcumin derivative ST09 effective against ovarian cancers. Prior in-vitro studies with ST09 drug showed cytotoxicity in tumorigenic cells compared to normal cells and in-vivo, significant tumor reduction was observed with least systemic toxicity. ST09 induced cytotoxicity in the ovarian cancer cells triggering mitochondria-mediated intrinsic apoptotic pathway. Delving deeper to understand the underlying molecular mechanisms involved in ovarian cancer pathogenesis, we identified an inverse correlation of miR-199a-5p with DDR1, a collagen receptor with receptor tyrosine kinase activity. The ST09 treatment in ovarian cancer cell lines resulted in the deregulation of the miR-199a-5p/DDR1 axis, conferring tumor-suppressive functions. We established DDR1 to be a direct target of miR-199a-5p and that ST09-induced DDR1 loss in these ovarian cancer cells resulted in the inactivation of its downstream MMP activation, migration, EMT, and prosurvival NF-κB pathway. Overall this study demonstrates ST09, a potent drug candidate for ovarian cancer treatment which exhibits anti-invasive and migrastatic properties.
Collapse
|
10
|
Fowler AJ, Ahn J, Hebron M, Chiu T, Ayoub R, Mulki S, Ressom H, Torres-Yaghi Y, Wilmarth B, Pagan FL, Moussa C. CSF MicroRNAs Reveal Impairment of Angiogenesis and Autophagy in Parkinson Disease. Neurol Genet 2021; 7:e633. [PMID: 34786477 PMCID: PMC8589263 DOI: 10.1212/nxg.0000000000000633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022]
Abstract
Background and Objectives We assessed longitudinal changes in CSF microRNAs (miRNAs) in patients with moderately severe Parkinson disease. Methods We used next-generation whole-genome miRNA sequencing to determine CSF miRNA expression in 75 patients with Parkinson disease after single random ascending doses of nilotinib and longitudinal miRNA expression after daily nilotinib, 150 and 300 mg, vs placebo for 1 year. Results Significant changes in the expression of miRNAs that control genes and pathways that regulate angiogenesis, autophagy, and the blood-brain-barrier components, primarily collagen, were observed over 1 year, suggesting impairment of these pathways in Parkinson progression in these patients. Different miRNAs that indicate activation of genes associated with autophagy flux and clearance and angiogenesis were significantly altered in the nilotinib, 300 mg vs 150 mg, or placebo group, and these changes correlated with clinical outcomes. No changes were observed in miRNAs after a single dose of nilotinib vs placebo. Discussion This study suggests vascular and autophagy defects in Parkinson progression. Nilotinib, 300 mg, reverses these effects via alteration of miRNA expression, suggesting epigenomic changes that may underlie long-term disease-modifying effects. Trial Registration Information Clinical trial registration number: NCT02954978.
Collapse
Affiliation(s)
- Alan J Fowler
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Jaeil Ahn
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Michaeline Hebron
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Timothy Chiu
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Reem Ayoub
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Sanjana Mulki
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Habtom Ressom
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Yasar Torres-Yaghi
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Barbara Wilmarth
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Fernando L Pagan
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| | - Charbel Moussa
- Translational Neurotherapeutics Program (A.J.F., M.H., T.C., R.A., S.M., B.W., F.L.P., C.M.), Department of Neurology; Interdisciplinary Program in Neuroscience (A.J.F.); Department of Biostatistics, Bioinformatics, and Biomathematics (J.A.); Department of Oncology (H.R.), Lombardi Comprehensive Cancer Center, Georgetown University Medical Center; and Movement Disorders Clinic (Y.T.Y., B.W., F.L.P., C.M.), Department of Neurology, MedStar Georgetown University Hospital, Washington, DC
| |
Collapse
|
11
|
Chen L, Kong X, Fang Y, Paunikar S, Wang X, Brown JAL, Bourke E, Li X, Wang J. Recent Advances in the Role of Discoidin Domain Receptor Tyrosine Kinase 1 and Discoidin Domain Receptor Tyrosine Kinase 2 in Breast and Ovarian Cancer. Front Cell Dev Biol 2021; 9:747314. [PMID: 34805157 PMCID: PMC8595330 DOI: 10.3389/fcell.2021.747314] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Discoidin domain receptor tyrosine kinases (DDRs) are a class of receptor tyrosine kinases (RTKs), and their dysregulation is associated with multiple diseases (including cancer, chronic inflammatory conditions, and fibrosis). The DDR family members (DDR1a-e and DDR2) are widely expressed, with predominant expression of DDR1 in epithelial cells and DDR2 in mesenchymal cells. Structurally, DDRs consist of three regions (an extracellular ligand binding domain, a transmembrane domain, and an intracellular region containing a kinase domain), with their kinase activity induced by receptor-specific ligand binding. Collagen binding to DDRs stimulates DDR phosphorylation activating kinase activity, signaling to MAPK, integrin, TGF-β, insulin receptor, and Notch signaling pathways. Abnormal DDR expression is detected in a range of solid tumors (including breast, ovarian, cervical liver, gastric, colorectal, lung, and brain). During tumorigenesis, abnormal activation of DDRs leads to invasion and metastasis, via dysregulation of cell adhesion, migration, proliferation, secretion of cytokines, and extracellular matrix remodeling. Differential expression or mutation of DDRs correlates with pathological classification, clinical characteristics, treatment response, and prognosis. Here, we discuss the discovery, structural characteristics, organizational distribution, and DDR-dependent signaling. Importantly, we highlight the key role of DDRs in the development and progression of breast and ovarian cancer.
Collapse
Affiliation(s)
- Li Chen
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shishir Paunikar
- Discipline of Pathology, School of Medicine, Lambe Institute for Translational Research, National University of Ireland Galway, Galway, Ireland
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - James A. L. Brown
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Health Research Institute, University of Limerick, Limerick, Ireland
| | - Emer Bourke
- Discipline of Pathology, School of Medicine, Lambe Institute for Translational Research, National University of Ireland Galway, Galway, Ireland
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Bonfil RD, Chen W, Vranic S, Sohail A, Shi D, Jang H, Kim HR, Prunotto M, Fridman R. Expression and subcellular localization of Discoidin Domain Receptor 1 (DDR1) define prostate cancer aggressiveness. Cancer Cell Int 2021; 21:507. [PMID: 34548097 PMCID: PMC8456559 DOI: 10.1186/s12935-021-02206-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The Discoidin Domain Receptor 1 (DDR1) is one of the two members of a unique family of receptor tyrosine kinase receptors that signal in response to collagen, which has been implicated in cancer progression. Here, we examined the expression of DDR1 in prostate cancer (PCa), and assessed its potential value as a prognostic marker, as a function of grade, stage and other clinicopathologic parameters. METHODS We investigated the association between the expression level and subcellular localization of DDR1 protein and PCa aggressiveness by immunohistochemistry, using tissue microarrays (TMAs) encompassing 200 cases of PCa with various Gleason scores (GS) and pathologic stages with matched normal tissue, and a highly specific monoclonal antibody. RESULTS DDR1 was found to be localized in the membrane, cytoplasm, and nuclear compartments of both normal and cancerous prostate epithelial cells. Analyses of DDR1 expression in low GS (≤ 7[3 + 4]) vs high GS (≥ 7[4 + 3]) tissues showed no differences in nuclear or cytoplasmic DDR1in either cancerous or adjacent normal tissue cores. However, relative to normal-matched tissue, the percentage of cases with higher membranous DDR1 expression was significantly lower in high vs. low GS cancers. Although nuclear localization of DDR1 was consistently detected in our tissue samples and also in cultured human PCa and normal prostate-derived cell lines, its presence in that site could not be associated with disease aggressiveness. No associations between DDR1 expression and overall survival or biochemical recurrence were found in this cohort of patients. CONCLUSION The data obtained through multivariate logistic regression model analysis suggest that the level of membranous DDR1 expression status may represent a potential biomarker of utility for better determination of PCa aggressiveness.
Collapse
Affiliation(s)
- R Daniel Bonfil
- Division of Pathology, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, 3200 S. University Drive, Terry Building # 1337, Fort Lauderdale, FL, 33328-2018, USA.
| | - Wei Chen
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Anjum Sohail
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Scott Hall #8200, 540 E. Canfield St, Detroit, MI, 48201, USA
| | - Dongping Shi
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Scott Hall #8200, 540 E. Canfield St, Detroit, MI, 48201, USA
| | - Hyejeong Jang
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Hyeong-Reh Kim
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Scott Hall #8200, 540 E. Canfield St, Detroit, MI, 48201, USA
| | - Marco Prunotto
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Rafael Fridman
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Scott Hall #8200, 540 E. Canfield St, Detroit, MI, 48201, USA.
| |
Collapse
|
13
|
Romayor I, Márquez J, Benedicto A, Herrero A, Arteta B, Olaso E. Tumor DDR1 deficiency reduces liver metastasis by colon carcinoma and impairs stromal reaction. Am J Physiol Gastrointest Liver Physiol 2021; 320:G1002-G1013. [PMID: 33851541 DOI: 10.1152/ajpgi.00078.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Tumor DDR1 acts as a key factor during the desmoplastic response surrounding hepatic colorectal metastasis. Hepatic sinusoidal cell-derived soluble factors stimulate tumor DDR1 activation. DDR1 modulates matrix remodeling to promote metastasis in the liver through the interaction with hepatic stromal cells, specifically liver sinusoidal endothelial cells and hepatic stellate cells.
Collapse
Affiliation(s)
- Irene Romayor
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Joana Márquez
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Aitor Benedicto
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Alba Herrero
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Beatriz Arteta
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Elvira Olaso
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
14
|
Xie X, He H, Zhang N, Wang X, Rui W, Xu D, Zhu Y. Overexpression of DDR1 Promotes Migration, Invasion, Though EMT-Related Molecule Expression and COL4A1/DDR1/MMP-2 Signaling Axis. Technol Cancer Res Treat 2020; 19:1533033820973277. [PMID: 33234027 PMCID: PMC7705183 DOI: 10.1177/1533033820973277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose: Discoidin domain receptor 1 (DDR1) belongs to a novel class of receptor tyrosine kinases. Previous evidence indicates that DDR1 overexpression promotes the aggressive growth of bladder cancer (BC) cells. This study aimed to investigate the molecular mechanisms by which DDR1 influences BC. Methods: DDR1 was transfected into human BC RT4 cells. DDR1, COL4A1, and MMP-2 expression in 30 BC tissues and paired adjacent tissues were examined by real-time polymerase chain reaction (RT-PCR) and immunohistochemistry. Transwell assays were conducted to determine cell migration and invasion. RT-PCR and western blot (WB) were also used to measure the DDR1, COL4A1, MMP-2, and EMT-related gene (ZEB1 and SLUG) expression in RT4 cells after DDR1 overexpression. Results: COL4A1 and MMP-2 interacted with DDR1 in the PPI network. RT-PCR and immunohistochemistry results showed that both mRNA and protein levels of DDR1 and COL4A1 were significantly increased in BC tissue, while the expression of MMP-2 was increased only at the mRNA level (P < 0.05). Overexpression of DDR1 in RT4 cells significantly promoted their migratory and invasive capabilities in vitro (P < 0.05). Moreover, overexpression of DDR1 in RT4 cells increased the mRNA and protein expression of ZEB1, SLUG, COL4A1, and MMP-2 (P < 0.01). DDR1-mediated migration and invasion of RT4 cells were reversed after COL4A1-siRNA treatment. Conclusion: DDR1 may be a potential therapeutic target in BC patients.
Collapse
Affiliation(s)
- Xin Xie
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongchao He
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ning Zhang
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaojing Wang
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenbin Rui
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Danfeng Xu
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Zhu
- Department of Urology, Ruijin Hospital, 56694Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Reger de Moura C, Prunotto M, Sohail A, Battistella M, Jouenne F, Marbach D, Lebbé C, Fridman R, Mourah S. Discoidin Domain Receptors in Melanoma: Potential Therapeutic Targets to Overcome MAPK Inhibitor Resistance. Front Oncol 2020; 10:1748. [PMID: 33014862 PMCID: PMC7516126 DOI: 10.3389/fonc.2020.01748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/04/2020] [Indexed: 01/04/2023] Open
Abstract
Melanoma is a highly malignant skin cancer with high propensity to metastasize and develop drug resistance, making it a difficult cancer to treat. Current therapies targeting BRAF (V600) mutations are initially effective, but eventually tumors overcome drug sensitivity and reoccur. This process is accomplished in part by reactivating alternate signaling networks that reinstate melanoma proliferative and survival capacity, mostly through reprogramming of receptor tyrosine kinase (RTK) signaling. Evidence indicates that the discoidin domain receptors (DDRs), a set of RTKs that signal in response to collagen, are part of the kinome network that confer drug resistance. We previously reported that DDR1 is expressed in melanomas, where it can promote tumor malignancy in mouse models of melanoma, and thus, DDR1 could be a promising target to overcome drug resistance. In this review, we summarize the current knowledge on DDRs in melanoma and their implication for therapy, with emphasis in resistance to MAPK inhibitors.
Collapse
Affiliation(s)
- Coralie Reger de Moura
- Laboratory of Pharmacogenomics, Hôpital Saint-Louis, AP-HP, Paris, France
- INSERM, UMR_S976, Université de Paris, Paris, France
| | - Marco Prunotto
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Anjum Sohail
- Department of Pathology, School of Medicine, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Maxime Battistella
- INSERM, UMR_S976, Université de Paris, Paris, France
- Department of Pathology, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Fanelie Jouenne
- Laboratory of Pharmacogenomics, Hôpital Saint-Louis, AP-HP, Paris, France
- INSERM, UMR_S976, Université de Paris, Paris, France
| | - Daniel Marbach
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Celeste Lebbé
- INSERM, UMR_S976, Université de Paris, Paris, France
- Department of Dermatology, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Rafael Fridman
- Department of Pathology, School of Medicine, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Samia Mourah
- Laboratory of Pharmacogenomics, Hôpital Saint-Louis, AP-HP, Paris, France
- INSERM, UMR_S976, Université de Paris, Paris, France
| |
Collapse
|
16
|
Lorusso G, Rüegg C, Kuonen F. Targeting the Extra-Cellular Matrix-Tumor Cell Crosstalk for Anti-Cancer Therapy: Emerging Alternatives to Integrin Inhibitors. Front Oncol 2020; 10:1231. [PMID: 32793493 PMCID: PMC7387567 DOI: 10.3389/fonc.2020.01231] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network composed of a multitude of different macromolecules. ECM components typically provide a supportive structure to the tissue and engender positional information and crosstalk with neighboring cells in a dynamic reciprocal manner, thereby regulating tissue development and homeostasis. During tumor progression, tumor cells commonly modify and hijack the surrounding ECM to sustain anchorage-dependent growth and survival, guide migration, store pro-tumorigenic cell-derived molecules and present them to enhance receptor activation. Thereby, ECM potentially supports tumor progression at various steps from initiation, to local growth, invasion, and systemic dissemination and ECM-tumor cells interactions have long been considered promising targets for cancer therapy. Integrins represent key surface receptors for the tumor cell to sense and interact with the ECM. Yet, attempts to therapeutically impinge on these interactions using integrin inhibitors have failed to deliver anticipated results, and integrin inhibitors are still missing in the emerging arsenal of drugs for targeted therapies. This paradox situation should urge the field to reconsider the role of integrins in cancer and their targeting, but also to envisage alternative strategies. Here, we review the therapeutic targets implicated in tumor cell adhesion to the ECM, whose inhibitors are currently in clinical trials and may offer alternatives to integrin inhibition.
Collapse
Affiliation(s)
- Girieca Lorusso
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Curzio Rüegg
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - François Kuonen
- Department of Dermatology and Venereology, Hôpital de Beaumont, Lausanne University Hospital Center, Lausanne, Switzerland
| |
Collapse
|
17
|
Targeting Discoidin Domain Receptor 1 (DDR1) Signaling and Its Crosstalk with β 1-integrin Emerges as a Key Factor for Breast Cancer Chemosensitization upon Collagen Type 1 Binding. Int J Mol Sci 2020; 21:ijms21144956. [PMID: 32668815 PMCID: PMC7404217 DOI: 10.3390/ijms21144956] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 12/23/2022] Open
Abstract
Collagen type 1 (COL1) is a ubiquitously existing extracellular matrix protein whose high density in breast tissue favors metastasis and chemoresistance. COL1-binding of MDA-MB-231 and MCF-7 breast cancer cells is mainly dependent on β1-integrins (ITGB1). Here, we elucidate the signaling of chemoresistance in both cell lines and their ITGB1-knockdown mutants and elucidated MAPK pathway to be strongly upregulated upon COL1 binding. Notably, Discoidin Domain Receptor 1 (DDR1) was identified as another important COL1-sensor, which is permanently active but takes over the role of COL1-receptor maintaining MAPK activation in ITGB1-knockdown cells. Consequently, inhibition of DDR1 and ERK1/2 act synergistically, and sensitize the cells for cytostatic treatments using mitoxantrone, or doxorubicin, which was associated with an impaired ABCG2 drug efflux transporter activity. These data favor DDR1 as a promising target for cancer cell sensitization, most likely in combination with MAPK pathway inhibitors to circumvent COL1 induced transporter resistance axis. Since ITGB1-knockdown also induces upregulation of pEGFR in MDA-MB-231 cells, inhibitory approaches including EGFR inhibitors, such as gefitinib appear promising for pharmacological interference. These findings provide evidence for the highly dynamic adaptation of breast cancer cells in maintaining matrix binding to circumvent cytotoxicity and highlight DDR1 signaling as a target for sensitization approaches.
Collapse
|
18
|
Chappell WH, Candido S, Abrams SL, Akula SM, Steelman LS, Martelli AM, Ratti S, Cocco L, Cervello M, Montalto G, Nicoletti F, Libra M, McCubrey JA. Influences of TP53 and the anti-aging DDR1 receptor in controlling Raf/MEK/ERK and PI3K/Akt expression and chemotherapeutic drug sensitivity in prostate cancer cell lines. Aging (Albany NY) 2020; 12:10194-10210. [PMID: 32492656 PMCID: PMC7346063 DOI: 10.18632/aging.103377] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/20/2020] [Indexed: 12/20/2022]
Abstract
Background: TP53 plays critical roles in sensitivity to chemotherapy, and aging. Collagen is very important in aging. The molecular structure and biochemical properties of collagen changes during aging. The discoidin domain receptor (DDR1) is regulated in part by collagen. Elucidating the links between TP53 and DDR1 in chemosensitivity and aging could improve therapies against cancer and aging. Results: Restoration of WT-TP53 activity resulted in increased sensitivity to chemotherapeutic drugs and elevated expression of key components of the Raf/MEK/ERK, PI3K/Akt and DDR1 pathways. DDR1 could modulate the levels of Raf/MEK/ERK and PI3K/Akt pathways as well as sensitize the cells to chemotherapeutic drugs. In contrast, suppression of WT TP53 with a dominant negative (DN) TP53 gene, suppressed DDR1 protein levels and increased their chemoresistance. Conclusion: Restoration of WT TP53 activity or increased expression of the anti-aging DDR1 collagen receptor can result in enhanced sensitivity to chemotherapeutic drugs. Our innovative studies indicate the important links between WT TP53 and DDR1 which can modulate Raf/MEK/ERK and PI3K/Akt signaling as well as chemosensitivity and aging. Methods: We investigated the roles of wild type (WT) and mutant TP53 on drug sensitivity of prostate cancer cells and the induction of Raf/MEK/ERK, PI3K/Akt and DDR1 expression and chemosensitivity.
Collapse
Affiliation(s)
- William H Chappell
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.,Current Address: Becton, Dickinson and Company (BD), BD Diagnostics, Franklin Lakes, NJ 07417, USA
| | - Saverio Candido
- Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Shaw M Akula
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Giuseppe Montalto
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy.,Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Massimo Libra
- Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
19
|
Discoidin Domain Receptor-1 (DDR1) is Involved in Angiolymphatic Invasion in Oral Cancer. Cancers (Basel) 2020; 12:cancers12040841. [PMID: 32244515 PMCID: PMC7226486 DOI: 10.3390/cancers12040841] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
The discoidin domain receptor-1 (DDR1) is a non-integrin collagen receptor recently implicated in the collective cell migration of other cancer types. Previously, we identified an elevated expression of DDR1 in oral squamous cell carcinoma (OSCC) cells. Through the data mining of a microarray dataset composed of matched tumor-normal tissues from forty OSCC patients, we distilled overexpressed genes statistically associated with angiolymphatic invasion, including DDR1, COL4A5, COL4A6 and PDPN. Dual immunohistochemical staining further confirmed the spatial locations of DDR1 and PDPN in OSCC tissues indicative of collective cancer cell invasion. An elevated DDR1 expression at both the transcription and protein level was observed by treating keratinocytes with collagen of fibrillar or basement membrane types. In addition, inhibition of DDR1 kinase activity in OSCC TW2.6 cells disrupted cell cohesiveness in a 2D culture, reduced spheroid invasion in a collagen gel matrix, and suppressed angiolymphatic invasion in xenograft tissues. Taken together, these results suggest that collagen deposition in the affected tissues followed by DDR1 overexpression could be central to OSCC tumor growth and angiolymphatic invasion. Thus, DDR1 inhibitors are potential therapeutic compounds in restraining oral cancer, which has not been previously explored.
Collapse
|
20
|
Tao Y, Wang R, Lai Q, Wu M, Wang Y, Jiang X, Zeng L, Zhou S, Li Z, Yang T, Yao Y, Wu Y, Yu L, Fu Y, Lai W, Peng Y, Lu Y, Zhang Z, Guo C, Zhang G, Gou L, Yang J. Targeting of DDR1 with antibody-drug conjugates has antitumor effects in a mouse model of colon carcinoma. Mol Oncol 2019; 13:1855-1873. [PMID: 31116512 PMCID: PMC6717758 DOI: 10.1002/1878-0261.12520] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/03/2019] [Accepted: 05/20/2019] [Indexed: 02/05/2023] Open
Abstract
DDR1 has been identified as a cancer‐associated receptor tyrosine kinase that is highly expressed in several malignancies relative to normal tissues. Clinically approved multi‐kinase inhibitors, such as nilotinib, inhibit DDR1‐mediated tumor growth in xenograft models, suggesting DDR1 might be a potential target for cancer treatments. Here, we employed an antibody‐based strategy with a novel anti‐DDR1 antibody‐drug conjugate (ADC) for colon carcinoma treatment. We developed T4H11‐DM4, an ADC targeting DDR1 which carries the tubulin inhibitor payload DM4. Immunohistochemical analysis of a tissue microarray containing 100 colon cancer specimens revealed that DDR1 was highly expressed in 81% of tumor tissues. Meanwhile, high expression of DDR1 was associated with poor survival in patients. In vitro, T4H11‐DM4 exhibited potent anti‐proliferative activity with half maximal inhibitory concentration (IC50) values in the nanomolar range in a panel of colon cancer cell lines. In vivo, the antitumor efficacy of T4H11‐DM4 was evaluated in three colon cancer cell lines expressing different levels of DDR1. T4H11‐DM4 achieved complete tumor regression at doses of 5 and 10 mg·kg−1 in HT‐29 and HCT116 tumor models. Moreover, a correlation between in vivo efficacy of T4H11‐DM4 and the levels of DDR1 expression on the cell surface was observed. Tumor cell proliferation was caused by the induction of mitotic arrest, indicating that the antitumor effect in vivo was mediated by DM4. In addition, T4H11‐DM4 was efficacious in oxaliplatin‐resistant colon cancer models. In exploratory safety studies, T4H11‐DM4 exhibited no overt toxicities when multi‐doses were administered at 10 mg·kg−1 into BALB/c nude mice or when a single dose up to 50 mg·kg−1 was administered into BALB/c mice. Overall, our findings highlight the potential of DDR1‐targeted ADC and may facilitate the development of a new effective therapeutic strategy for colon cancer.
Collapse
Affiliation(s)
- Yiran Tao
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ruixue Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qinhuai Lai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Mengdan Wu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohua Jiang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lishi Zeng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shijie Zhou
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongping Li
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tinghan Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuqin Yao
- West China School of Public Health and Healthy Food Evaluation Research Center/No. 4 West China Teaching Hospital, Sichuan University, Chengdu, China
| | - Yangping Wu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Yu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, China
| | - Yuyin Fu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weirong Lai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yujia Peng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Lu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhixiong Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Cuiyu Guo
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Guangbing Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lantu Gou
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, China
| |
Collapse
|
21
|
Ankney JA, Xie L, Wrobel JA, Wang L, Chen X. Novel secretome-to-transcriptome integrated or secreto-transcriptomic approach to reveal liquid biopsy biomarkers for predicting individualized prognosis of breast cancer patients. BMC Med Genomics 2019; 12:78. [PMID: 31146747 PMCID: PMC6543675 DOI: 10.1186/s12920-019-0530-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 05/13/2019] [Indexed: 02/08/2023] Open
Abstract
Background Presently, a 50-gene expression model (PAM50) serves as a breast cancer (BC) subtype classifier that is insufficient to distinguish, within each single PAM50-classified subtype, patient subpopulations having different prognosis. There is a pressing need for inexpensive and minimally invasive biomarker tests to easily and accurately predict individuals’ clinical outcomes and response to treatments. Although quantitative proteomic approaches have been developed to identify/profile proteins secreted (secretome) from various cancer cell lines in vitro, missing are the clinicopathological relevance and the associated prognostic value of these secretomic identifications. Methods To discover biomarkers to predict individualized prognosis we introduce a new multi-omics (secreto-transcriptomics) method that identifies, in their oncogenically secreted states, candidate markers of BC subtypes whose genes bear patient-specific mRNA expression alterations of prognostic significance. First, we used label-free quantitative (LFQ) proteomics to identify the proteins showing BC-subtypic secretion from a series of BC cell lines representing major BC-subtypes. To determine and externally validate the prognostic value of these secreted proteins, we developed a secreto-transcriptomic approach that discovered a PAM50-subtypic Secretion-Correlated mRNA Expression Pattern (SeCEP) wherein the PAM50-subtypic secretion of select proteins statistically correlated with cis-mRNA expression of their encoding genes in patients of the corresponding PAM50-subtypes. Kaplan-Meier analysis of SeCEP genes was used to identify new liquid biopsy biomarkers for predicting individualized prognosis. Results The mRNA expression-to-secretion correlation (SeCEP) pinpointed multiple genes that are fully translated into the oncogenically active secretome in a PAM50-subtypic manner. Further, multiple SeCEP genes in distinct combinations or panels of multiple SeCEP genes were identified as ‘systems prognostic markers’ that showed mRNA co-overexpression patterns in the distinct subpopulations of PAM50-subtypic patients with poor prognosis or high-risk of relapse. Thus, our secreto-transcriptomic approach statistically linked BC subtypic secretome genes with patient-specific information about their mRNA expression alterations and significantly improved the sensitivity and specificity in patient stratification in the context of clinical outcomes or prognosis. Conclusions By combining LFQ secretome screening with proteo-transcriptomic retrospective analysis of patient data our integrated multi-omics approach bypasses costly, tedious, genome-wide fishing and predictive modeling that are commonly required to distinguish a few prognostically altered genes from thousands of other non-BC related genes in a genome. Electronic supplementary material The online version of this article (10.1186/s12920-019-0530-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J Astor Ankney
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ling Xie
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - John A Wrobel
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Li Wang
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xian Chen
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
22
|
Zhao J, Ji D, Zhai X, Zhang L, Luo X, Fu X. Oral Administration of Prunella vulgaris L Improves the Effect of Taxane on Preventing the Progression of Breast Cancer and Reduces Its Side Effects. Front Pharmacol 2018; 9:806. [PMID: 30123125 PMCID: PMC6085460 DOI: 10.3389/fphar.2018.00806] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022] Open
Abstract
We aimed to explore the efficacy and safety of Prunella vulgaris L (PVL) combined with taxane for treatment of patients with breast cancer (BC). The main ingredients of PVL were analyzed by high-performance liquid chromatography (HPLC). In the experiment, 424 patients with BC were evenly assigned into two groups: experimental group (EG, oral administration of PVL and taxane) and control group (CG, oral administration of placebo and taxane). The primary endpoint was pathologic complete response (pCR), which was evaluated using Miller and Payne system. The secondary endpoints included adverse events (AE) and overall survival (OS), which were evaluated by Common Terminology Criteria for Adverse Event version and Kaplan-Meier curves, respectively. Response Evaluation Criteria in Solid Tumors was used to evaluate the clinical efficacy of PVL. Estrogen receptor (ER) status was also measured. The main side effects were compared between the two groups. The main ingredients of PVL were caffeic acid and rosmarinic acid, which both exert anti-tumor properties. The average follow-up time was 41 months. Eighteen and 31 patients dropped out from EG and CG, respectively. Overall, pCRs were detected in 94 cases (25.1%), comprising 61 cases (31.4%) from EG and 33 cases (18.2%) from CG (P < 0.05). PVL treatment improved the pCR rate and OS time compared with those in CG (P < 0.05). The 3-year OS rates were 86.5 and 77.2% in patients from EG and CG, respectively (P < 0.05). Moreover, ER status was associated with pCR rate and could be an independent prognostic factor in BC. Moreover, treatment with PVL prevented side effects, namely, neutrophil-reduced fever and anemia caused by chemotherapy. Hence, chemotherapy using PVL and taxane could be a safe and effective treatment for patients with BC. PVL may be a potential adjuvant medicine for BC treatment.
Collapse
Affiliation(s)
- Jixue Zhao
- Department of Pediatric Surgery, The First Hospital of Jilin University, Changchun, China
| | - Degang Ji
- Department of Hepatobiliary Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xujie Zhai
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lirong Zhang
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiao Luo
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xin Fu
- Department of Nursing, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|