1
|
Yin G, Li N, Chen X, Zhang Y, Fang J, Zhong Q, Huang Z, Guo W. Gene mutations and differentiation in laryngeal and pharyngeal squamous cell carcinoma. Discov Oncol 2025; 16:624. [PMID: 40293562 PMCID: PMC12037971 DOI: 10.1007/s12672-025-02407-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
OBJECTIVE To explore the characteristics of genomic variation patterns in Chinese patients with laryngeal and pharyngeal squamous cell carcinoma (SCC) and their correlation with differentiation and clinical significance. METHODS We analyzed genomic variations in 45 patients. Mutation patterns were evaluated using the 688 panel. We evaluated the correlation among degree of differentiation, patient prognosis, and mutation status and also analyzed 564 HNSCC samples from the UALCAN database. RESULTS Significant differences were observed in overall survival (OS) and progression-free survival (PFS) among patients with different degrees of differentiation. Based on the DriverML model, we found that the genes with the highest mutation rates were neurogenic locus notch homolog protein 1 (NOTCH1), tumor protein 53 (TP53), FAT atypical cadherin 1 (FAT1), and mitogen-activated protein kinase kinase kinase 4 (MAP3 K4) (over 30%). We are the first to our knowledge to propose that MAP3 K4 (33%) may be a driving gene for Chinese SCC patients. Moreover, NOTCH1 and CUB and sushi multiple domains 3 (CSMD3) were mutually exclusive (p < 0.05). CSMD3 mutations were primarily found in poorly differentiated patients (83%, 5/6). Furthermore, NOTCH1wild and MAP3 K4wild were mainly present in poorly differentiated patients (p = 0.011) as well. We also validated the differential expression of NOTCH1 and MAP3 K4 and their association (p < 0.05) with tumor differentiation using 564 HNSCC samples from the UALCAN database. CONCLUSION We identified a potential new driving gene, MAP3 K4, in Chinese SCC patients and confirmed that the interaction between NOTCH1-MAP3 K4 may affect the differentiation of laryngeal and pharyngeal SCC. However, further exploration and large-scale sample validation are needed.
Collapse
Affiliation(s)
- Gaofei Yin
- Otolaryngology head and neck surgery, Beijing Tongren Hospital, Capital Medical University, No 1. Dongjiaomin Lane,Dongcheng District, Beijing, 100730, China
| | - Nuan Li
- Otolaryngology head and neck surgery, Beijing Tongren Hospital, Capital Medical University, No 1. Dongjiaomin Lane,Dongcheng District, Beijing, 100730, China
| | - Xiaohong Chen
- Otolaryngology head and neck surgery, Beijing Tongren Hospital, Capital Medical University, No 1. Dongjiaomin Lane,Dongcheng District, Beijing, 100730, China
| | - Yang Zhang
- Otolaryngology head and neck surgery, Beijing Tongren Hospital, Capital Medical University, No 1. Dongjiaomin Lane,Dongcheng District, Beijing, 100730, China
| | - Jugao Fang
- Otolaryngology head and neck surgery, Beijing Tongren Hospital, Capital Medical University, No 1. Dongjiaomin Lane,Dongcheng District, Beijing, 100730, China
| | - Qi Zhong
- Otolaryngology head and neck surgery, Beijing Tongren Hospital, Capital Medical University, No 1. Dongjiaomin Lane,Dongcheng District, Beijing, 100730, China
| | - Zhigang Huang
- Otolaryngology head and neck surgery, Beijing Tongren Hospital, Capital Medical University, No 1. Dongjiaomin Lane,Dongcheng District, Beijing, 100730, China.
| | - Wei Guo
- Otolaryngology head and neck surgery, Beijing Tongren Hospital, Capital Medical University, No 1. Dongjiaomin Lane,Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
2
|
Nose K, Taniguchi K, Fujita M, Moyama C, Mori M, Ishita M, Yoshida T, Ii H, Sakai T, Nakata S. γ-Glutamylcyclotransferase is transcriptionally regulated by c-Jun and controls proliferation of glioblastoma stem cells through Notch1 levels. Cancer Gene Ther 2024; 31:1831-1839. [PMID: 39394529 DOI: 10.1038/s41417-024-00835-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
Glioblastoma stem cells (GSCs) have been reported to cause poor prognosis of glioblastoma by contributing to therapy resistance. γ-Glutamylcyclotransferase (GGCT) is highly expressed in various cancer types, including glioblastoma, and its inhibition suppresses cancer cell growth. However, the mechanism of GGCT overexpression and its function in GSCs are unknown. In this study, we show that GGCT is highly expressed in GSCs established from a mouse glioblastoma model and its knockdown suppresses their proliferation. Effects of NRas and its downstream transcription factor c-Jun on GGCT expression were analyzed; NRas knockdown reduced c-Jun and GGCT expression. Knockdown of c-Jun also reduced expression levels of GGCT and inhibited cell proliferation. Consistent with this, pharmacological inhibition of c-Jun with SP600125 reduced GGCT and inhibited GSC proliferation. Furthermore, the GGCT promoter-reporter assay with mutagenesis demonstrated that c-Jun regulates the activity of the GGCT promoter via AP-1 consensus sequence. Gene expression analysis revealed that GGCT knockdown showed a repressive effect on the Delta-Notch pathway and decreased Notch1 expression. Notch1 knockdown alone inhibited the GSC proliferation, confirming that Notch1 is functional in this model. Forced expression of the Notch1 intracellular domain restored the growth inhibitory effect of GGCT knockdown. Moreover, GGCT knockdown inhibited GSC tumorigenic potential in vivo. These results indicate that GGCT, whose expression is promoted by c-Jun, plays an important role in the proliferation and tumorigenic potential of GSCs, and that the phenotype caused by its knockdown is contributed by a decrease in Notch1. Thus, GGCT may represent a novel therapeutic target for attacking GSCs.
Collapse
Affiliation(s)
- Kozue Nose
- Laboratory of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Keiko Taniguchi
- Department of Drug Discovery Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mitsugu Fujita
- Center for Medical Education and Clinical Training, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Chiami Moyama
- Laboratory of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan
- Department of Pharmacy, Shiga University of Medical Science Hospital, Shiga, Japan
| | - Masaya Mori
- Laboratory of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Mizuki Ishita
- Laboratory of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tatsushi Yoshida
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiromi Ii
- Laboratory of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Toshiyuki Sakai
- Department of Drug Discovery Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Susumu Nakata
- Laboratory of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan.
| |
Collapse
|
3
|
Abdel-Mohsen MA, Badawy AM, Abu-Youssef MA, Yehia MA, Abou Shamaa LD, Mohamed SA. Influence of copper(I) nicotinate complex on the Notch1 signaling pathway in triple negative breast cancer cell lines. Sci Rep 2024; 14:2522. [PMID: 38291201 PMCID: PMC10827744 DOI: 10.1038/s41598-024-52952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/25/2024] [Indexed: 02/01/2024] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer which is characterized by its aggressiveness, poor and short overall survival. In this concept, there is a growing demand for metal-based compounds in TNBC therapy as copper complex that have a less toxic effect on normal cells and could stimulate apoptotic cell death. Additionally, Notch1 signaling pathway has received great attention as one of the most important potential targets for developing a novel therapeutic strategy. The present study is an attempt to assess the promising chemotherapeutic activities of copper(I) nicotinate (CNC) through its impact on the expression of downstream genes of Notch1 signaling pathway and the cell fate of TNBC. The co-treatment of TNBC cells with doxorubicin (Doxo) and CNC was also investigated. To approach the objective of the present study, TNBC cell lines; HCC1806 and MDAMB231, were utilized. MTT assay was used to determine the IC50 values of CNC and Doxo. After treatment, microtubule-associated protein light chain3 (LC3) were determined by flow cytometry. Additionally, qRT-PCR technique was used to detect the changes in genes levels that are involved Notch1 signaling pathway. Moreover, autophagosomes were monitored and imaged by Transmission electron microscopy. Treatment of TNBC cells with CNC modulated Notch1 signaling pathway in different manners with respect to the type of cells and the applied dose of CNC. The observed effects of CNC may reflect the possible anti-cancer activities of CNC in both types of TNBC. However, cell type and CNC dose should be considered.
Collapse
Affiliation(s)
- Mohamed A Abdel-Mohsen
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt.
| | - Asmaa M Badawy
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt.
| | - Morsy A Abu-Youssef
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mona A Yehia
- Histochemistry and Cell Biology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Lobna D Abou Shamaa
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Shymaa Abdullah Mohamed
- Molecular Biology Unit, Medical Technology Center and Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
4
|
Kumari L, Mishra L, Patel P, Sharma N, Gupta GD, Kurmi BD. Emerging targeted therapeutic strategies for the treatment of triple-negative breast cancer. J Drug Target 2023; 31:889-907. [PMID: 37539789 DOI: 10.1080/1061186x.2023.2245579] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023]
Abstract
Triple-negative breast cancer (TNBC), a subtype of breast cancer that lacks expression of oestrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER-2), has clinical features including a high degree of invasiveness, an elevated risk of metastasis, tendency to relapse, and poor prognosis. It constitutes around 10-15% of all breast cancer, and having heredity of BRCA1 mutated breast cancer could be a reason for the occurrence of TNBC in women. Overexpression of cellular and molecular targets, i.e. CD44 receptor, EGFR receptor, Folate receptor, Transferrin receptor, VEGF receptor, and Androgen receptor, have emerged as promising targets for treating TNBC. Signalling pathways such as Notch signalling and PI3K/AKT/mTOR also play a significant role in carrying out and managing crucial pro-survival and pro-growth cellular processes that can be utilised for targeted therapy against triple-negative breast cancer. This review sheds light on various targeting strategies, including cellular and molecular targets, signalling pathways, poly (ADP-ribose) polymerase inhibitors, antibody-drug conjugates, and immune checkpoint inhibitors PARP, immunotherapy, ADCs have all found a place in the current TNBC therapeutic paradigm. The role of photothermal therapy (PTT) and photodynamic therapy (PDT) has also been explored briefly.
Collapse
Affiliation(s)
- Lakshmi Kumari
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| | - Lopamudra Mishra
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, Moga, Punjab, India
| | - Nitin Sharma
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| | | | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| |
Collapse
|
5
|
Hossain F, Ucar DA, Monticone G, Ran Y, Majumder S, Larter K, Luu H, Wyczechowska D, Heidari S, Xu K, Shanthalingam S, Matossian M, Xi Y, Burow M, Collins-Burow B, Del Valle L, Hicks C, Zabaleta J, Golde T, Osborne B, Miele L. Sulindac sulfide as a non-immune suppressive γ-secretase modulator to target triple-negative breast cancer. Front Immunol 2023; 14:1244159. [PMID: 37901240 PMCID: PMC10612326 DOI: 10.3389/fimmu.2023.1244159] [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: 06/21/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) comprises a heterogeneous group of clinically aggressive tumors with high risk of recurrence and metastasis. Current pharmacological treatment options remain largely limited to chemotherapy. Despite promising results, the efficacy of immunotherapy and chemo-immunotherapy in TNBC remains limited. There is strong evidence supporting the involvement of Notch signaling in TNBC progression. Expression of Notch1 and its ligand Jagged1 correlate with poor prognosis. Notch inhibitors, including g-secretase inhibitors (GSIs), are quite effective in preclinical models of TNBC. However, the success of GSIs in clinical trials has been limited by their intestinal toxicity and potential for adverse immunological effects, since Notch plays key roles in T-cell activation, including CD8 T-cells in tumors. Our overarching goal is to replace GSIs with agents that lack their systemic toxicity and ideally, do not affect tumor immunity. We identified sulindac sulfide (SS), the active metabolite of FDA-approved NSAID sulindac, as a potential candidate to replace GSIs. Methods We investigated the pharmacological and immunotherapeutic properties of SS in TNBC models in vitro, ex-vivo and in vivo. Results We confirmed that SS, a known γ-secretase modulator (GSM), inhibits Notch1 cleavage in TNBC cells. SS significantly inhibited mammosphere growth in all human and murine TNBC models tested. In a transplantable mouse TNBC tumor model (C0321), SS had remarkable single-agent anti-tumor activity and eliminated Notch1 protein expression in tumors. Importantly, SS did not inhibit Notch cleavage in T- cells, and the anti-tumor effects of SS were significantly enhanced when combined with a-PD1 immunotherapy in our TNBC organoids and in vivo. Discussion Our data support further investigation of SS for the treatment of TNBC, in conjunction with chemo- or -chemo-immunotherapy. Repurposing an FDA-approved, safe agent for the treatment of TNBC may be a cost-effective, rapidly deployable therapeutic option for a patient population in need of more effective therapies.
Collapse
Affiliation(s)
- Fokhrul Hossain
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Deniz A. Ucar
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Giulia Monticone
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Yong Ran
- Department of Pharmacological and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Kristina Larter
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Hanh Luu
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Dorota Wyczechowska
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
| | - Soroor Heidari
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Keli Xu
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Sudarvili Shanthalingam
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | | | - Yaguang Xi
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Matthew Burow
- School of Medicine, Tulane University, New Orleans, LA, United States
| | | | - Luis Del Valle
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
- Department of Pathology, Louisiana State University Health Sciences Center - New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Chindo Hicks
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
| | - Todd Golde
- Department of Pharmacological and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Barbara Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| |
Collapse
|
6
|
Sen P, Ghosh SS. The Intricate Notch Signaling Dynamics in Therapeutic Realms of Cancer. ACS Pharmacol Transl Sci 2023; 6:651-670. [PMID: 37200816 PMCID: PMC10186364 DOI: 10.1021/acsptsci.2c00239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Indexed: 05/20/2023]
Abstract
The Notch pathway is remarkably simple without the interventions of secondary messengers. It possesses a unique receptor-ligand interaction that imparts signaling upon cleavage of the receptor followed by the nuclear localization of its cleaved intracellular domain. It is found that the transcriptional regulator of the Notch pathway lies at the intersection of multiple signaling pathways that enhance the aggressiveness of cancer. The preclinical and clinical evidence supports the pro-oncogenic function of Notch signaling in various tumor subtypes. Owing to its oncogenic role, the Notch signaling pathway assists in enhanced tumorigenesis by facilitating angiogenesis, drug resistance, epithelial to mesenchymal transition, etc., which is also attributed to the poor outcome in patients. Therefore, it is extremely vital to discover a suitable inhibitor to downregulate the signal-transducing ability of Notch. The Notch inhibitory agents, such as receptor decoys, protease (ADAM and γ-secretase) inhibitors, and monoclonal/bispecific antibodies, are being investigated as candidate therapeutic agents. Studies conducted by our group exemplify the promising results in ablating tumorigenic aggressiveness by inhibiting the constituents of the Notch pathway. This review deals with the detailed mechanism of the Notch pathways and their implications in various malignancies. It also bestows us with the recent therapeutic advances concerning Notch signaling in the context of monotherapy and combination therapy.
Collapse
Affiliation(s)
- Plaboni Sen
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Siddhartha Sankar Ghosh
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Centre
for Nanotechnology, Indian Institute of
Technology Guwahati, Guwahati 781039, Assam, India
| |
Collapse
|
7
|
Sen P, Kandasamy T, Ghosh SS. In-silico evidence of ADAM metalloproteinase pathology in cancer signaling networks. J Biomol Struct Dyn 2022; 40:11771-11786. [PMID: 34402747 DOI: 10.1080/07391102.2021.1964602] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lack of effective targeted therapies often contributes to poor clinical outcomes of aggressive malignancies associated with drug resistance, angiogenesis and metastasis. Literature mining portrays the major role of ADAM17 in cancer and inflammatory diseases. However, it is quite challenging to design a candidate drug for targeting ADAM17 due to its structural similarity with the catalytic domain of the matrix metalloproteases (MMPs). The present study reports the protein-protein interaction analysis of ADAM17, along with the molecular docking and MD simulation studies for the screened compounds. Our analysis confirms the association of ADAM17 with numerous oncogenes that facilitates cancer progression and inflammation, especially the members of the Notch, receptor tyrosine kinase (RTK) and TNFα pathways. The outcome provides evidence that the prevalent protease ADAM17 could attribute to cancer signaling regulation though the shedding of various inflammatory and oncogenic molecules. We have also exploited the analogues of the existing inhibitors, with an aim at discovering a potent molecule, which could be repurposed as a drug against ADAM17 inflicted cancer progression. Upon stringent screening, we delineated our choice into two specific compounds (I6 and I9; analogues of IK862, a type of y-lactam hydroxamates), possessing the lowest binding energy (-9.1 Kcal/mol), stable MD-simulation studies and superior pharmacodynamic properties. The current information illustrates the avenue to persuade further research on targeting ADAM17 with small molecular compounds (I6 and I9) in cancer therapeutics.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Plaboni Sen
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Thirukumaran Kandasamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.,Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|
8
|
Herbal Ingredients in the Prevention of Breast Cancer: Comprehensive Review of Potential Molecular Targets and Role of Natural Products. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6044640. [PMID: 36017236 PMCID: PMC9398845 DOI: 10.1155/2022/6044640] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/06/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022]
Abstract
Among various cancers, breast cancer is the most prevalent type in women throughout the world. Breast cancer treatment is challenging due to complex nature of the etiology of disease. Cell division cycle alterations are often encountered in a variety of cancer types including breast cancer. Common treatments include chemotherapy, surgery, radiotherapy, and hormonal therapy; however, adverse effects and multidrug resistance lead to complications and noncompliance. Accordingly, there is an increasing demand for natural products from medicinal plants and foods. This review summarizes molecular mechanisms of signaling pathways in breast cancer and identifies mechanisms by which natural compounds may exert their efficacy in the treatment of breast cancer.
Collapse
|
9
|
Chimento A, D’Amico M, Pezzi V, De Amicis F. Notch Signaling in Breast Tumor Microenvironment as Mediator of Drug Resistance. Int J Mol Sci 2022; 23:6296. [PMID: 35682974 PMCID: PMC9181656 DOI: 10.3390/ijms23116296] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 01/10/2023] Open
Abstract
Notch signaling dysregulation encourages breast cancer progression through different mechanisms such as stem cell maintenance, cell proliferation and migration/invasion. Furthermore, Notch is a crucial driver regulating juxtracrine and paracrine communications between tumor and stroma. The complex interplay between the abnormal Notch pathway orchestrating the activation of other signals and cellular heterogeneity contribute towards remodeling of the tumor microenvironment. These changes, together with tumor evolution and treatment pressure, drive breast cancer drug resistance. Preclinical studies have shown that targeting the Notch pathway can prevent or reverse resistance, reducing or eliminating breast cancer stem cells. In the present review, we will summarize the current scientific evidence that highlights the involvement of Notch activation within the breast tumor microenvironment, angiogenesis, extracellular matrix remodeling, and tumor/stroma/immune system interplay and its involvement in mechanisms of therapy resistance.
Collapse
Affiliation(s)
- Adele Chimento
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (A.C.); (M.D.); (F.D.A.)
| | - Maria D’Amico
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (A.C.); (M.D.); (F.D.A.)
- Health Center, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Vincenzo Pezzi
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (A.C.); (M.D.); (F.D.A.)
| | - Francesca De Amicis
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (A.C.); (M.D.); (F.D.A.)
- Health Center, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| |
Collapse
|
10
|
Lal R, Ritchie J, Richmond L, Keeshan K. Detecting endogenous TRIB2 protein expression by flow cytometry and Western blotting. Methods Enzymol 2022; 667:59-77. [PMID: 35525555 DOI: 10.1016/bs.mie.2022.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Protein kinases catalyze the transfer of a phosphate group thereby activating proteins and initiating signaling cascades. Their cousins, the pseudokinases, are enzymatically nonactive counterparts of protein kinases that can be considered zombie enzymes. Interestingly, pseudokinases, which constitute about 10% of the human kinome, have been implicated in many cancers, despite their sequences predicting a lack of catalytic activity. Owing to recent research, it has been demonstrated that dysregulation of many pseudokinases triggers changes in cell signaling, proliferation, and drug resistance. This review is aimed at describing methods that can be used for detection of Tribbles family of pseudokinases, specifically TRIB2. We describe intracellular staining by flow cytometry and Western blotting techniques for the detection of endogenous TRIB2 protein.
Collapse
Affiliation(s)
- Ridhima Lal
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, SC, United Kingdom
| | - Jake Ritchie
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, SC, United Kingdom
| | - Laura Richmond
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, SC, United Kingdom
| | - Karen Keeshan
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, SC, United Kingdom.
| |
Collapse
|
11
|
The mevalonate pathway in breast cancer biology. Cancer Lett 2022; 542:215761. [DOI: 10.1016/j.canlet.2022.215761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/07/2023]
|
12
|
Naranjo AI, González-Gómez MJ, Baladrón V, Laborda J, Nueda ML. Different Expression Levels of DLK2 Inhibit NOTCH Signaling and Inversely Modulate MDA-MB-231 Breast Cancer Tumor Growth In Vivo. Int J Mol Sci 2022; 23:1554. [PMID: 35163478 PMCID: PMC8836183 DOI: 10.3390/ijms23031554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 11/22/2022] Open
Abstract
NOTCH signaling is implicated in the development of breast cancer tumors. DLK2, a non-canonical inhibitor of NOTCH signaling, was previously shown to be involved in skin and breast cancer. In this work, we studied whether different levels of DLK2 expression influenced the breast cancer characteristics of MDA-MB-231 cells. We found that DLK2 overexpression inhibited NOTCH activation in a dose-dependent manner. Moreover, depending on the level of inhibition of NOTCH1 activation generated by different levels of DLK2 expression, cell proliferation, cell cycle dynamics, cell apoptosis, cell migration, and tumor growth in vivo were affected in opposite directions. Low levels of DLK2 expression produced a slight inhibition of NOTCH1 activation, and enhanced MDA-MB-231 cell invasion in vitro and cell proliferation both in vitro and in vivo. In contrast, MDA-MB-231 cells expressing elevated levels of DLK2 showed a strong inhibition of NOTCH1 activation, decreased cell proliferation, increased cell apoptosis, and were unable to generate tumors in vivo. In addition, DLK2 expression levels also affected some members of other cell signaling pathways implicated in cancer, such as ERK1/2 MAPK, AKT, and rpS6 kinases. Our data support an important role of DLK2 as a protein that can finely regulate NOTCH signaling and affect the tumor properties and growth dynamics of MDA-MB-231 breast cancer cells.
Collapse
Affiliation(s)
- Ana-Isabel Naranjo
- Biochemistry and Molecular Biology Branch, Medical School/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain; (A.-I.N.); (V.B.)
| | - María-Julia González-Gómez
- Biochemistry and Molecular Biology Branch, Higher Technical School of Agricultural and Forestry Engineering/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain;
| | - Victoriano Baladrón
- Biochemistry and Molecular Biology Branch, Medical School/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain; (A.-I.N.); (V.B.)
| | - Jorge Laborda
- Biochemistry and Molecular Biology Branch, School of Pharmacy/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain
| | - María-Luisa Nueda
- Biochemistry and Molecular Biology Branch, School of Pharmacy/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain
| |
Collapse
|
13
|
Hounjet J, Vooijs M. The Role of Intracellular Trafficking of Notch Receptors in Ligand-Independent Notch Activation. Biomolecules 2021; 11:biom11091369. [PMID: 34572582 PMCID: PMC8466058 DOI: 10.3390/biom11091369] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022] Open
Abstract
Aberrant Notch signaling has been found in a broad range of human malignancies. Consequently, small molecule inhibitors and antibodies targeting Notch signaling in human cancers have been developed and tested; however, these have failed due to limited anti-tumor efficacy because of dose-limiting toxicities in normal tissues. Therefore, there is an unmet need to discover novel regulators of malignant Notch signaling, which do not affect Notch signaling in healthy tissues. This review provides a comprehensive overview of the current knowledge on the role of intracellular trafficking in ligand-independent Notch receptor activation, the possible mechanisms involved, and possible therapeutic opportunities for inhibitors of intracellular trafficking in Notch targeting.
Collapse
|
14
|
Qayoom H, Wani NA, Alshehri B, Mir MA. An insight into the cancer stem cell survival pathways involved in chemoresistance in triple-negative breast cancer. Future Oncol 2021; 17:4185-4206. [PMID: 34342489 DOI: 10.2217/fon-2021-0172] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most complex, aggressive and fatal subtype of breast cancer. Owing to the lack of targeted therapy and heterogenic nature of TNBC, chemotherapy remains the sole treatment option for TNBC, with taxanes and anthracyclines representing the general chemotherapeutic regimen in TNBC therapy. But unfortunately, patients develop resistance to the existing chemotherapeutic regimen, resulting in approximately 90% treatment failure. Breast cancer stem cells (BCSCs) are one of the major causes for the development of chemoresistance in TNBC patients. After surviving the chemotherapy damage, the presence of BCSCs results in relapse and recurrence of TNBC. Several pathways are known to regulate BCSCs' survival, such as the Wnt/β-catenin, Hedgehog, JAK/STAT and HIPPO pathways. Therefore it is imperative to target these pathways in the context of eliminating chemoresistance. In this review we will discuss the novel strategies and various preclinical and clinical studies to give an insight into overcoming TNBC chemoresistance. We present a detailed account of recent studies carried out that open an exciting perspective in relation to the mechanisms of chemoresistance.
Collapse
Affiliation(s)
- Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, J&K, India
| | - Nissar A Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir Nunar Ganderbal 191201, J&K, India
| | - Bader Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, KSA
| | - Manzoor A Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, J&K, India
| |
Collapse
|
15
|
Edwards A, Brennan K. Notch Signalling in Breast Development and Cancer. Front Cell Dev Biol 2021; 9:692173. [PMID: 34295896 PMCID: PMC8290365 DOI: 10.3389/fcell.2021.692173] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/07/2021] [Indexed: 12/22/2022] Open
Abstract
The Notch signalling pathway is a highly conserved developmental signalling pathway, with vital roles in determining cell fate during embryonic development and tissue homeostasis. Aberrant Notch signalling has been implicated in many disease pathologies, including cancer. In this review, we will outline the mechanism and regulation of the Notch signalling pathway. We will also outline the role Notch signalling plays in normal mammary gland development and how Notch signalling is implicated in breast cancer tumorigenesis and progression. We will cover how Notch signalling controls several different hallmarks of cancer within epithelial cells with sections focussed on its roles in proliferation, apoptosis, invasion, and metastasis. We will provide evidence for Notch signalling in the breast cancer stem cell phenotype, which also has implications for therapy resistance and disease relapse in breast cancer patients. Finally, we will summarise the developments in therapeutic targeting of Notch signalling, and the pros and cons of this approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Abigail Edwards
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Keith Brennan
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
16
|
Fodor A, Lazar AL, Buchman C, Tiperciuc B, Orasan OH, Cozma A. MicroRNAs: The Link between the Metabolic Syndrome and Oncogenesis. Int J Mol Sci 2021; 22:ijms22126337. [PMID: 34199293 PMCID: PMC8231835 DOI: 10.3390/ijms22126337] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic syndrome (MetS) represents a cluster of disorders that increase the risk of a plethora of conditions, in particular type two diabetes, cardiovascular diseases, and certain types of cancers. MetS is a complex entity characterized by a chronic inflammatory state that implies dysregulations of adipokins and proinflammatory cytokins together with hormonal and growth factors imbalances. Of great interest is the implication of microRNA (miRNA, miR), non-coding RNA, in cancer genesis, progression, and metastasis. The adipose tissue serves as an important source of miRs, which represent a novel class of adipokines, that play a crucial role in carcinogenesis. Altered miRs secretion in the adipose tissue, in the context of MetS, might explain their implication in the oncogenesis. The interplay between miRs expressed in adipose tissue, their dysregulation and cancer pathogenesis are still intriguing, taking into consideration the fact that miRNAs show both carcinogenic and tumor suppressor effects. The aim of our review was to discuss the latest publications concerning the implication of miRs dysregulation in MetS and their significance in tumoral signaling pathways. Furthermore, we emphasized the role of miRNAs as potential target therapies and their implication in cancer progression and metastasis.
Collapse
Affiliation(s)
- Adriana Fodor
- Department of Diabetes and Nutrtion, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence: (A.F.); (A.L.L.); (C.B.)
| | - Andrada Luciana Lazar
- Department of Dermatology, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence: (A.F.); (A.L.L.); (C.B.)
| | - Cristina Buchman
- Department of Oncology, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence: (A.F.); (A.L.L.); (C.B.)
| | - Brandusa Tiperciuc
- Department of Pharmaceutical Chemistry, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Olga Hilda Orasan
- Internal Medicine Department, 4th Medical Clinic “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (O.H.O.); (A.C.)
| | - Angela Cozma
- Internal Medicine Department, 4th Medical Clinic “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (O.H.O.); (A.C.)
| |
Collapse
|
17
|
Porcelli L, Mazzotta A, Garofoli M, Di Fonte R, Guida G, Guida M, Tommasi S, Azzariti A. Active notch protects MAPK activated melanoma cell lines from MEK inhibitor cobimetinib. Biomed Pharmacother 2020; 133:111006. [PMID: 33202284 DOI: 10.1016/j.biopha.2020.111006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/26/2020] [Accepted: 11/08/2020] [Indexed: 12/18/2022] Open
Abstract
The crosstalk between Notch and MAPK pathway plays a role in MEK inhibitor resistance in BRAFV600E metastatic melanoma (MM) and promotes migration in GNAQQ209L uveal melanoma (UM) cells. We determined the cytotoxicity of combinatorial inhibition of MEK and Notch by cobimetinib and γ-secretase inhibitor (GSI) nirogacestat, in BRAFV600E and BRAF wt MM and GNAQQ209L UM cells displaying different Erk1/2 and Notch activation status, with the aim to elucidate the impact of Notch signaling in the response to MEK inhibitor. Overall the combination was synergic in BRAFV600E MM and GNAQQ209L UM cells and antagonistic in BRAF wt one. Focusing on UM cells, we found that cobimetinib resulted in G0/G1 phase arrest and apoptosis induction, whereas the combination with GSI increased treatment efficacy by inducing a senescent-like state of cells and by blocking migration towards liver cancer cells. Mechanistically, this was reflected in a strong reduction of cyclin D1, in the inactivation of retinoblastoma protein and in the increase of p27KIP1 expression levels. Of note, each drug alone prevented Notch signaling activation resulting in inhibition of c-jun(Ser63) and Hes-1 expression. The combination achieved the strongest inhibition on Notch signaling and on both c-jun(Ser63) and Erk1/2 activation level. In conclusion we unveiled a coordinate action of MAPK and Notch signaling in promoting proliferation of BRAFV600E MM and GNAQQ209L UM cells. Remarkably, the simultaneous inhibition of MEK and Notch signaling highlighted a role for the second pathway in protecting cells against senescence in GNAQQ209L UM cells treated with the MEK inhibitor.
Collapse
Affiliation(s)
- Letizia Porcelli
- Experimental Pharmacology Laboratory, Italia, 70124, Bari, Italy
| | | | | | - Roberta Di Fonte
- Experimental Pharmacology Laboratory, Italia, 70124, Bari, Italy
| | - Gabriella Guida
- Department of Basic Medical Sciences Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy
| | | | - Stefania Tommasi
- Molecular Diagnostics and Pharmacogenetics Unit IRCCS Istituto Tumori "Giovanni Paolo II" di Bari, Italia, 70124, Bari, Italy
| | - Amalia Azzariti
- Experimental Pharmacology Laboratory, Italia, 70124, Bari, Italy.
| |
Collapse
|
18
|
Gharaibeh L, Elmadany N, Alwosaibai K, Alshaer W. Notch1 in Cancer Therapy: Possible Clinical Implications and Challenges. Mol Pharmacol 2020; 98:559-576. [PMID: 32913140 DOI: 10.1124/molpharm.120.000006] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022] Open
Abstract
The Notch family consists of four highly conserved transmembrane receptors. The release of the active intracellular domain requires the enzymatic activity of γ-secretase. Notch is involved in embryonic development and in many physiologic processes of normal cells, in which it regulates growth, apoptosis, and differentiation. Notch1, a member of the Notch family, is implicated in many types of cancer, including breast cancer (especially triple-negative breast cancer), leukemias, brain tumors, and many others. Notch1 is tightly connected to many signaling pathways that are therapeutically involved in tumorigenesis. Together, they impact apoptosis, proliferation, chemosensitivity, immune response, and the population of cancer stem cells. Notch1 inhibition can be achieved through various and diverse methods, the most common of which are the γ-secretase inhibitors, which produce a pan-Notch inhibition, or the use of Notch1 short interference RNA or Notch1 monoclonal antibodies, which produce a more specific blockade. Downregulation of Notch1 can be used alone or in combination with chemotherapy, which can achieve a synergistic effect and a decrease in chemoresistance. Targeting Notch1 in cancers that harbor high expression levels of Notch1 offers an addition to therapeutic strategies recruited for managing cancer. Considering available evidence, Notch1 offers a legitimate target that might be incorporated in future strategies for combating cancer. In this review, the possible clinical applications of Notch1 inhibition and the obstacles that hinder its clinical application are discussed. SIGNIFICANCE STATEMENT: Notch1 plays an important role in different types of cancer. Numerous approaches of Notch1 inhibition possess potential benefits in the management of various clinical aspects of cancer. The application of different Notch1 inhibition modalities faces many challenges.
Collapse
Affiliation(s)
- L Gharaibeh
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan (L.G); Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.E.); Research Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia (K.A.); and Cell Therapy Center, The University of Jordan, Amman, Jordan (W.A.)
| | - N Elmadany
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan (L.G); Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.E.); Research Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia (K.A.); and Cell Therapy Center, The University of Jordan, Amman, Jordan (W.A.)
| | - K Alwosaibai
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan (L.G); Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.E.); Research Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia (K.A.); and Cell Therapy Center, The University of Jordan, Amman, Jordan (W.A.)
| | - W Alshaer
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan (L.G); Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.E.); Research Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia (K.A.); and Cell Therapy Center, The University of Jordan, Amman, Jordan (W.A.)
| |
Collapse
|
19
|
Zhao Y, Zeng D, Wang H, Sun N, Xin J, Yang H, Lei L, Khalique A, Rajput DS, Pan K, Shu G, Jing B, Ni X. Analysis of miRNA Expression in the Ileum of Broiler Chickens During Bacillus licheniformis H2 Supplementation Against Subclinical Necrotic Enteritis. Probiotics Antimicrob Proteins 2020; 13:356-366. [PMID: 32975724 DOI: 10.1007/s12602-020-09709-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 01/20/2023]
Abstract
Subclinical necrotic enteritis (SNE) is one of the serious threats to the poultry industry. Probiotics have been proven to exert beneficial effects in controlling SNE. However, their exact mechanisms have not been fully elucidated. Moreover, few studies have focused on their impact on microRNAs (miRNAs). Therefore, the present study aimed to explore the miRNA expression profiles in the ileum of broiler chickens during probiotic supplementation for controlling SNE. A total of 180 newly hatched male broilers were randomly allocated into three groups, including a negative control group, an SNE infection group, and a Bacillus licheniformis H2 pretreatment group. Illumina high-throughput sequencing was conducted to identify the miRNA expression of the three groups. Results showed that 628 miRNAs, including 582 known miRNAs and 46 novel miRNAs, were detected in the miRNA libraries. The target genes of 57 significantly differentially expressed miRNAs were predicted and annotated. Moreover, they were found to be partly enriched in pathways related to immunity and inflammation such as tumor necrosis factor receptor binding, immune response-regulating signaling pathway, Toll-like receptor 2 signaling pathway, interleukin-15 production, activation of NF-κB-inducing kinase activity, and MAP kinase tyrosine/serine/threonine phosphatase activity. Some of the target genes of 57 miRNAs were related to the MAPK signaling pathway. Furthermore, the expression of several miRNAs, which may be involved in the MAPK signaling pathway, was significantly affected by SNE induction and showed no significant difference in the presence of H2. All these findings provide comprehensive miRNA expression profiles of three different treatment groups. They further suggest that H2 could exert beneficial effects in controlling SNE through immune and inflammatory response associated with altered miRNA expression, such as the MAPK signaling pathway.
Collapse
Affiliation(s)
- Ying Zhao
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hesong Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jinge Xin
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hanbo Yang
- Chengdu Slan Biotechnology Co., Ltd, Chengdu, China
| | - Lei Lei
- Chengdu Slan Biotechnology Co., Ltd, Chengdu, China
| | - Abdul Khalique
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Danish Sharafat Rajput
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gang Shu
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
20
|
Liubomirski Y, Ben-Baruch A. Notch-Inflammation Networks in Regulation of Breast Cancer Progression. Cells 2020; 9:cells9071576. [PMID: 32605277 PMCID: PMC7407628 DOI: 10.3390/cells9071576] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/21/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Members of the Notch family and chronic inflammation were each separately demonstrated to have prominent malignancy-supporting roles in breast cancer. Recent investigations indicate that bi-directional interactions that exist between these two pathways promote the malignancy phenotype of breast tumor cells and of their tumor microenvironment. In this review article, we demonstrate the importance of Notch-inflammation interplays in malignancy by describing three key networks that act in breast cancer and their impacts on functions that contribute to disease progression: (1) Cross-talks of the Notch pathway with myeloid cells that are important players in cancer-related inflammation, focusing mainly on macrophages; (2) Cross-talks of the Notch pathway with pro-inflammatory factors, exemplified mainly by Notch interactions with interleukin 6 and its downstream pathways (STAT3); (3) Cross-talks of the Notch pathway with typical inflammatory transcription factors, primarily NF-κB. These three networks enhance tumor-promoting functions in different breast tumor subtypes and act in reciprocal manners, whereby Notch family members activate inflammatory elements and vice versa. These characteristics illustrate the fundamental roles played by Notch-inflammation interactions in elevating breast cancer progression and propose that joint targeting of both pathways together may provide more effective and less toxic treatment approaches in this disease.
Collapse
|
21
|
Cholesterol and beyond - The role of the mevalonate pathway in cancer biology. Biochim Biophys Acta Rev Cancer 2020; 1873:188351. [PMID: 32007596 DOI: 10.1016/j.bbcan.2020.188351] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/14/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023]
Abstract
Cancer is a multifaceted global disease. Transformation of a normal to a malignant cell takes several steps, including somatic mutations, epigenetic alterations, metabolic reprogramming and loss of cell growth control. Recently, the mevalonate pathway has emerged as a crucial regulator of tumor biology and a potential therapeutic target. This pathway controls cholesterol production and posttranslational modifications of Rho-GTPases, both of which are linked to several key steps of tumor progression. Inhibitors of the mevalonate pathway induce pleiotropic antitumor-effects in several human malignancies, identifying the pathway as an attractive candidate for novel therapies. In this review, we will provide an overview about the role and regulation of the mevalonate pathway in certain aspects of cancer initiation and progression and its potential for therapeutic intervention in oncology.
Collapse
|
22
|
Krishna BM, Jana S, Singhal J, Horne D, Awasthi S, Salgia R, Singhal SS. Notch signaling in breast cancer: From pathway analysis to therapy. Cancer Lett 2019; 461:123-131. [PMID: 31326555 DOI: 10.1016/j.canlet.2019.07.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 01/15/2023]
Abstract
The Notch signaling pathway, which is highly conserved from sea urchins to humans, plays an important role in cell-differentiation, survival, proliferation, stem-cell renewal, and determining cell fate during development and morphogenesis. It is well established that signaling pathways are dysregulated in a wide-range of diseases, including human malignancies. Studies suggest that the dysregulation of the Notch pathway contributes to carcinogenesis, cancer stem cell renewal, angiogenesis, and chemo-resistance. Elevated levels of Notch receptors and ligands have been associated with cancer-progression and poor survival. Furthermore, the Notch signaling pathway regulates the transcriptional activity of key target genes through crosstalk with several other signaling pathways. Indeed, increasing evidence suggests that the Notch signaling pathway may serve as a therapeutic target for the treatment of several cancers, including breast cancer. Researchers have demonstrated the anti-tumor properties of Notch inhibitors in various cancer types. Currently, Notch inhibitors are being evaluated for anticancer efficacy in a number of clinical-trials. However, because there are multiple Notch receptors that can exhibit either oncogenic or tumor-suppressing roles in various cells, it is important that the Notch inhibitors are specific to particular receptors that are tumorigenic in nature. This review critically evaluates existing Notch inhibitory drugs and strategies and summarizes the previous discoveries, current understandings, and recent developments in support of Notch receptors as therapeutic targets in breast cancer.
Collapse
Affiliation(s)
- B Madhu Krishna
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - Samir Jana
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - Jyotsana Singhal
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - Sanjay Awasthi
- Department of Internal Medicine, Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Ravi Salgia
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - Sharad S Singhal
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA.
| |
Collapse
|
23
|
Notch Signaling Activation as a Hallmark for Triple-Negative Breast Cancer Subtype. JOURNAL OF ONCOLOGY 2019; 2019:8707053. [PMID: 31379945 PMCID: PMC6657611 DOI: 10.1155/2019/8707053] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/19/2019] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) is a subgroup of 15%-20% of diagnosed breast cancer patients. It is generally considered to be the most difficult breast cancer subtype to deal with, due to the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), which usually direct targeted therapies. In this scenario, the current treatments of TNBC-affected patients rely on tumor excision and conventional chemotherapy. As a result, the prognosis is overall poor. Thus, the identification and characterization of targets for novel therapies are urgently required. The Notch signaling pathway has emerged to act in the pathogenesis and tumor progression of TNBCs. Firstly, Notch receptors are associated with the regulation of tumor-initiating cells (TICs) behavior, as well as with the aetiology of TNBCs. Secondly, there is a strong evidence that Notch pathway is a relevant player in mammary cancer stem cells maintenance and expansion. Finally, Notch receptors expression and activation strongly correlate with the aggressive clinicopathological and biological phenotypes of breast cancer (e.g., invasiveness and chemoresistance), which are relevant characteristics of TNBC subtype. The purpose of this up-to-date review is to provide a detailed overview of the specific role of all four Notch receptors (Notch1, Notch2, Notch3, and Notch4) in TNBCs, thus identifying the Notch signaling pathway deregulation/activation as a pathognomonic feature of this breast cancer subtype. Furthermore, this review will also discuss recent information associated with different therapeutic options related to the four Notch receptors, which may be useful to evaluate prognostic or predictive indicators as well as to develop new therapies aimed at improving the clinical outcome of TNBC patients.
Collapse
|
24
|
Singh S, Chakrabarti R. Consequences of EMT-Driven Changes in the Immune Microenvironment of Breast Cancer and Therapeutic Response of Cancer Cells. J Clin Med 2019; 8:jcm8050642. [PMID: 31075939 PMCID: PMC6572359 DOI: 10.3390/jcm8050642] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/30/2019] [Accepted: 05/04/2019] [Indexed: 02/06/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a process through which epithelial cells lose their epithelial characteristics and cell–cell contact, thus increasing their invasive potential. In addition to its well-known roles in embryonic development, wound healing, and regeneration, EMT plays an important role in tumor progression and metastatic invasion. In breast cancer, EMT both increases the migratory capacity and invasive potential of tumor cells, and initiates protumorigenic alterations in the tumor microenvironment (TME). In particular, recent evidence has linked increased expression of EMT markers such as TWIST1 and MMPs in breast tumors with increased immune infiltration in the TME. These immune cells then provide cues that promote immune evasion by tumor cells, which is associated with enhanced tumor progression and metastasis. In the current review, we will summarize the current knowledge of the role of EMT in the biology of different subtypes of breast cancer. We will further explore the correlation between genetic switches leading to EMT and EMT-induced alterations within the TME that drive tumor growth and metastasis, as well as their possible effect on therapeutic response in breast cancer.
Collapse
Affiliation(s)
- Snahlata Singh
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Rumela Chakrabarti
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
Liubomirski Y, Lerrer S, Meshel T, Morein D, Rubinstein-Achiasaf L, Sprinzak D, Wiemann S, Körner C, Ehrlich M, Ben-Baruch A. Notch-Mediated Tumor-Stroma-Inflammation Networks Promote Invasive Properties and CXCL8 Expression in Triple-Negative Breast Cancer. Front Immunol 2019; 10:804. [PMID: 31105691 PMCID: PMC6492532 DOI: 10.3389/fimmu.2019.00804] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 03/26/2019] [Indexed: 01/12/2023] Open
Abstract
Stromal cells and pro-inflammatory cytokines play key roles in promoting the aggressiveness of triple-negative breast cancers (TNBC; Basal/Basal-like). In our previous study we demonstrated that stimulation of TNBC and mesenchymal stem cells (MSCs) co-cultures by the pro-inflammatory cytokine tumor necrosis factor α (TNFα) has led to increased metastasis-related properties in vitro and in vivo. In this context, elevated release of the pro-metastatic chemokines CXCL8 (IL-8) and CCL5 (RANTES) was noted in TNFα- and interleukin-1β (IL-1β)-stimulated TNBC:MSC co-cultures; the process was partly (CXCL8) and entirely (CCL5) dependent on physical contacts between the two cell types. Here, we demonstrate that DAPT, inhibitor of γ-secretase that participates in activation of Notch receptors, inhibited the migration and invasion of TNBC cells that were grown in “Contact” co-cultures with MSCs or with patient-derived cancer-associated fibroblasts (CAFs), in the presence of TNFα. DAPT also inhibited the contact-dependent induction of CXCL8, but not of CCL5, in TNFα- and IL-1β-stimulated TNBC:MSC/CAF co-cultures; some level of heterogeneity between the responses of different TNBC cell lines was noted, with MDA-MB-231:MSC/CAF co-cultures being the most sensitive to DAPT. Patient dataset studies comparing basal tumors to luminal-A tumors, and mRNA analyses of Notch receptors in TNBC and luminal-A cells pointed at Notch1 as possible mediator of CXCL8 increase in TNFα-stimulated TNBC:stroma “Contact” co-cultures. Accordingly, down-regulation of Notch1 in TNBC cells by siRNA has substantially reduced the contact-dependent elevation in CXCL8 in TNFα- and also in IL-1β-stimulated TNBC:MSC “Contact” co-cultures. Then, studies in which CXCL8 or p65 (NF-κB pathway) were down-regulated (siRNAs; CRISPR/Cas9) in TNBC cells and/or MSCs, indicated that upon TNFα stimulation of “Contact” co-cultures, p65 was activated and led to CXCL8 production mainly in TNBC cells. Moreover, our findings indicated that when tumor cells interacted with stromal cells in the presence of pro-inflammatory stimuli, TNFα-induced p65 activation has led to elevated Notch1 expression and activation, which then gave rise to elevated production of CXCL8. Overall, tumor:stroma interactions set the stage for Notch1 activation by pro-inflammatory signals, leading to CXCL8 induction and consequently to pro-metastatic activities. These observations may have important clinical implications in designing novel therapy combinations in TNBC.
Collapse
Affiliation(s)
- Yulia Liubomirski
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shalom Lerrer
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tsipi Meshel
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Dina Morein
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Linor Rubinstein-Achiasaf
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - David Sprinzak
- School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cindy Körner
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcelo Ehrlich
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adit Ben-Baruch
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
26
|
Jing L, Ruan Z, Sun H, Li Q, Han L, Huang L, Yu S, Wang Y, Guo H, Jiao M. Epithelial-mesenchymal transition induced cancer-stem-cell-like characteristics in hepatocellular carcinoma. J Cell Physiol 2019; 234:18448-18458. [PMID: 30908631 DOI: 10.1002/jcp.28480] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma in China accounts for half of the world's incidence. Both epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs) are thought to be involved in tumor malignant progression. However, the relationship between EMT and CSCs is still unclear. Bioinformatics analysis was performed to evaluate the relationship between EMT and CSCs. The EMT and CSC regulatory mechanism was investigated through Transwell, wound-healing, sphere formation, colony-forming, and western blotting assays. Immunofluorescence and immunoprecipitation were used to study the interaction of hypoxia inducible factor 1α (HIF-1α) /Notch1. Immunohistochemical study was applied to investigate the expression pattern in the process of hepatocellular carcinogenesis and development. In our present study, bioinformatics results indicate that the expression of EMT-related molecules is correlated with CSCs. In vitro studies indicated that EMT activation could induce CSC characteristics. Notch1 was confirmed to mediate the process of EMT-induced CSCs through the interaction with HIF-1α directly. Our findings indicate that EMT could induce CSC-like characteristics, which is mediated by HIF-1α-upregulated Notch intracellular domain expression.
Collapse
Affiliation(s)
- Li Jing
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhiping Ruan
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haifeng Sun
- The Third Department of Medical Oncology, Shaanxi Provincial Tumor Hospital, Xi'an, China
| | - Qing Li
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lili Han
- Department of Oncology, he Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lanxuan Huang
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Sizhe Yu
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yu Wang
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hui Guo
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Min Jiao
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
27
|
Rajarajan D, Selvarajan S, Charan Raja MR, Kar Mahapatra S, Kasiappan R. Genome-wide analysis reveals miR-3184-5p and miR-181c-3p as a critical regulator for adipocytes-associated breast cancer. J Cell Physiol 2019; 234:17959-17974. [PMID: 30847933 DOI: 10.1002/jcp.28428] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/03/2019] [Accepted: 02/14/2019] [Indexed: 12/14/2022]
Abstract
Obesity is considered as an independent risk factor for breast cancer (BCa) and plays a major role in the breast tumor microenvironment. The etiology and mechanisms by which obesity contributes to BCa development is not yet understood. Herein, we show that in vitro coculture of BCa cells with mature adipocytes (MA-BCa) increased proliferation, migration, and invasive phenotype of BCa cells. MA-BCa coculture led to increased production of proinflammatory cytokines and chemokines. To identify microRNAs (miRNAs) in BCa cells that are modulated by the presence of adipocytes, we used small RNA sequencing analysis. Sequencing data revealed that 98 miRNAs were differentially expressed in MA-BCa. Among them, miR-3184-5p and miR-181c-3p were found to be the most upregulated and downregulated miRNAs, and direct targets are FOXP4 and PPARα, respectively. In vitro functional assays using a combination of miR-3184-5p inhibitor and miR-181c-3p mimic synergistically decreased adipocytes-induced cell proliferation and invasive capacity of BCa cells. Gene Set Enrichment analysis indicated that transcription factors were highly enriched followed by protein kinases, oncogene, and protein regulators in MA-BCa. GeneGo Metacore pathway analysis uncovered "NOTCH-induced EMT pathway" was found to be the most abundant in MA-BCa. Consistently, epithelial-mesenchymal transition-associated markers were also increased in MA-BCa. The disease enrichment analysis of the predict target genes revealed that diabetes mellitus was significantly affected disease in MA-BCa. Taken together, our data suggest that miRNA-based regulatory mechanism associated with deregulation of pathways and biological functions orchestrated by adipocytes-secreted factors might drive the BCa progression and metastasis in obese patients.
Collapse
Affiliation(s)
- Dheeran Rajarajan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sweetha Selvarajan
- Department of Biochemistry, The Graduate Centre of the City University of New York (CUNY), New York
| | - Mamilla R Charan Raja
- Department of Biotechnology, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed To Be University, Thanjavur, India
| | - Santanu Kar Mahapatra
- Department of Biotechnology, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed To Be University, Thanjavur, India
| | - Ravi Kasiappan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
28
|
Otoukesh B, Boddouhi B, Moghtadaei M, Kaghazian P, Kaghazian M. Novel molecular insights and new therapeutic strategies in osteosarcoma. Cancer Cell Int 2018; 18:158. [PMID: 30349420 PMCID: PMC6192346 DOI: 10.1186/s12935-018-0654-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/01/2018] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS) is one of the most prevalent malignant cancers with lower survival and poor overall prognosis mainly in children and adolescents. Identifying the molecular mechanisms and OS stem cells (OSCs) as new concepts involved in disease pathogenesis and progression may potentially lead to new therapeutic targets. Therefore, therapeutic targeting of OSCs can be one of the most important and effective strategies for the treatment of OS. This review describes the new molecular targets of OS as well as novel therapeutic approaches in the design of future investigations and treatment.
Collapse
Affiliation(s)
- Babak Otoukesh
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, 1445613131 Iran
| | - Bahram Boddouhi
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, 1445613131 Iran
| | - Mehdi Moghtadaei
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, 1445613131 Iran
| | - Peyman Kaghazian
- Department of Orthopedic and Traumatology, Universitätsklinikum Bonn, Bonn, Germany
| | - Maria Kaghazian
- Department of Biology, Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
29
|
Positive feedback loop between mitochondrial fission and Notch signaling promotes survivin-mediated survival of TNBC cells. Cell Death Dis 2018; 9:1050. [PMID: 30323195 PMCID: PMC6189045 DOI: 10.1038/s41419-018-1083-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 07/31/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022]
Abstract
Mitochondrial morphology is remodeled by continuous dynamic cycles of fission and fusion. Emerging data have shown that the disturbance of balance between mitochondrial fission and fusion is involved in the progression of several types of neoplasms. However, the status of mitochondrial dynamics and its potential biological roles in breast cancer (BC), particularly in triple negative BC (TNBC) are not fully clear. Here, we reported that the mitochondrial fission was significantly increased in BC tissues, especially in the TNBC tissues, when compared with that in the corresponding peritumor tissues. Meanwhile, our data showed that Drp1 was upregulated, while Mfn1 was downregulated in TNBC. Moreover, elevated mitochondrial fission was associated with poorer prognosis in TNBC patients. Mitochondrial fission promoted the survival of TNBC cells both in vitro and in vivo. Furthermore, we identified a positive feedback loop between mitochondrial fission and Notch signaling pathway in TNBC cells, as proved by the experimental evidence that the activation of Notch signaling enhanced Drp1-mediated mitochondrial fission and Drp1-mediated mitochondrial fission in turn promoted the activation of Notch signaling, which ultimately promoted the cell survival of TNBC via increasing survivin expression level. Inhibition of either Notch1 or Drp1 significantly impaired the activation of the other, leading to the suppression of TNBC cell survival and proliferation. Collectively, our data reveal a novel mechanism that the positive feedback loop between mitochondrial fission and Notch signaling promotes the survival, proliferation and apoptotic resistance of TNBC cells via increasing survivin expression and thus favors cancer progression.
Collapse
|
30
|
Leontovich AA, Jalalirad M, Salisbury JL, Mills L, Haddox C, Schroeder M, Tuma A, Guicciardi ME, Zammataro L, Gambino MW, Amato A, Di Leonardo A, McCubrey J, Lange CA, Liu M, Haddad T, Goetz M, Boughey J, Sarkaria J, Wang L, Ingle JN, Galanis E, D'Assoro AB. NOTCH3 expression is linked to breast cancer seeding and distant metastasis. Breast Cancer Res 2018; 20:105. [PMID: 30180881 PMCID: PMC6123953 DOI: 10.1186/s13058-018-1020-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/12/2018] [Indexed: 02/08/2023] Open
Abstract
Background Development of distant metastases involves a complex multistep biological process termed the invasion-metastasis cascade, which includes dissemination of cancer cells from the primary tumor to secondary organs. NOTCH developmental signaling plays a critical role in promoting epithelial-to-mesenchymal transition, tumor stemness, and metastasis. Although all four NOTCH receptors show oncogenic properties, the unique role of each of these receptors in the sequential stepwise events that typify the invasion-metastasis cascade remains elusive. Methods We have established metastatic xenografts expressing high endogenous levels of NOTCH3 using estrogen receptor alpha-positive (ERα+) MCF-7 breast cancer cells with constitutive active Raf-1/mitogen-associated protein kinase (MAPK) signaling (vMCF-7Raf-1) and MDA-MB-231 triple-negative breast cancer (TNBC) cells. The critical role of NOTCH3 in inducing an invasive phenotype and poor outcome was corroborated in unique TNBC cells resulting from a patient-derived brain metastasis (TNBC-M25) and in publicly available claudin-low breast tumor specimens collected from participants in the Molecular Taxonomy of Breast Cancer International Consortium database. Results In this study, we identified an association between NOTCH3 expression and development of metastases in ERα+ and TNBC models. ERα+ breast tumor xenografts with a constitutive active Raf-1/MAPK signaling developed spontaneous lung metastases through the clonal expansion of cancer cells expressing a NOTCH3 reprogramming network. Abrogation of NOTCH3 expression significantly reduced the self-renewal and invasive capacity of ex vivo breast cancer cells, restoring a luminal CD44low/CD24high/ERαhigh phenotype. Forced expression of the mitotic Aurora kinase A (AURKA), which promotes breast cancer metastases, failed to restore the invasive capacity of NOTCH3-null cells, demonstrating that NOTCH3 expression is required for an invasive phenotype. Likewise, pharmacologic inhibition of NOTCH signaling also impaired TNBC cell seeding and metastatic growth. Significantly, the role of aberrant NOTCH3 expression in promoting tumor self-renewal, invasiveness, and poor outcome was corroborated in unique TNBC cells from a patient-derived brain metastasis and in publicly available claudin-low breast tumor specimens. Conclusions These findings demonstrate the key role of NOTCH3 oncogenic signaling in the genesis of breast cancer metastasis and provide a compelling preclinical rationale for the design of novel therapeutic strategies that will selectively target NOTCH3 to halt metastatic seeding and to improve the clinical outcomes of patients with breast cancer. Electronic supplementary material The online version of this article (10.1186/s13058-018-1020-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexey A Leontovich
- Department of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Mohammad Jalalirad
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Jeffrey L Salisbury
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Lisa Mills
- Department of Molecular Medicine, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Candace Haddox
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Mark Schroeder
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Ann Tuma
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Maria E Guicciardi
- Department of Internal Medicine, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Luca Zammataro
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Mario W Gambino
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Angela Amato
- Department of Cellular and Developmental Biology, University of Palermo, Palermo, Italy
| | - Aldo Di Leonardo
- Department of Cellular and Developmental Biology, University of Palermo, Palermo, Italy
| | - James McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Carol A Lange
- Department of Medicine and Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Minetta Liu
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Tufia Haddad
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Matthew Goetz
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Judy Boughey
- Department of Surgery, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Jann Sarkaria
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Liewei Wang
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - James N Ingle
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Evanthia Galanis
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA.,Department of Molecular Medicine, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Antonino B D'Assoro
- Department of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA. .,Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA.
| |
Collapse
|
31
|
Intratumor MAPK and PI3K signaling pathway heterogeneity in glioblastoma tissue correlates with CREB signaling and distinct target gene signatures. Exp Mol Pathol 2018; 105:23-31. [DOI: 10.1016/j.yexmp.2018.05.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 05/26/2018] [Indexed: 11/20/2022]
|
32
|
Notch-1-PTEN-ERK1/2 signaling axis promotes HER2+ breast cancer cell proliferation and stem cell survival. Oncogene 2018; 37:4489-4504. [PMID: 29743588 DOI: 10.1038/s41388-018-0251-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 03/14/2018] [Accepted: 03/16/2018] [Indexed: 12/13/2022]
Abstract
Trastuzumab targets the HER2 receptor on breast cancer cells to attenuate HER2-driven tumor growth. However, resistance to trastuzumab-based therapy remains a major clinical problem for women with HER2+ breast cancer. Breast cancer stem cells (BCSCs) are suggested to be responsible for drug resistance and tumor recurrence. Notch signaling has been shown to promote BCSC survival and self-renewal. Trastuzumab-resistant cells have increased Notch-1 expression. Notch signaling drives cell proliferation in vitro and is required for tumor recurrence in vivo. We demonstrate herein a mechanism by which Notch-1 is required for trastuzumab resistance by repressing PTEN expression to contribute to activation of ERK1/2 signaling. Furthermore, Notch-1-mediated inhibition of PTEN is necessary for BCSC survival in vitro and in vivo. Inhibition of MEK1/2-ERK1/2 signaling in trastuzumab-resistant breast cancer cells mimics effects of Notch-1 knockdown on bulk cell proliferation and BCSC survival. These findings suggest that Notch-1 contributes to trastuzumab resistance by repressing PTEN and this may lead to hyperactivation of ERK1/2 signaling. Furthermore, high Notch-1 and low PTEN mRNA expression may predict poorer overall survival in women with breast cancer. Notch-1 protein expression predicts poorer survival in women with HER2+ breast cancer. These results support a potential future clinical trial combining anti-Notch-1 and anti-MEK/ERK therapy for trastuzumab-resistant breast cancer.
Collapse
|
33
|
Ahn JS, Ann EJ, Kim MY, Yoon JH, Lee HJ, Jo EH, Lee K, Lee JS, Park HS. Autophagy negatively regulates tumor cell proliferation through phosphorylation dependent degradation of the Notch1 intracellular domain. Oncotarget 2018; 7:79047-79063. [PMID: 27806347 PMCID: PMC5346697 DOI: 10.18632/oncotarget.12986] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 10/19/2016] [Indexed: 02/01/2023] Open
Abstract
Autophagy is a highly conserved mechanism that degrades long-lived proteins and dysfunctional organelles, and contributes to cell fate. In this study, autophagy attenuates Notch1 signaling by degrading the Notch1 intracellular domain (Notch1-IC). Nutrient-deprivation promotes Notch1-IC phosphorylation by MEKK1 and phosphorylated Notch1-IC is recognized by Fbw7 E3 ligase. The ubiquitination of Notch1-IC by Fbw7 is essential for the interaction between Notch1-IC and p62 and for the formation of aggregates. Inhibition of Notch1 signaling prevents the transformation of breast cancer cells, tumor progression, and metastasis. The expression of Notch1 and p62 is inversely correlated with Beclin1 expression in human breast cancer patients. These results show that autophagy inhibits Notch1 signaling by promoting Notch1-IC degradation and therefore plays a role in tumor suppression.
Collapse
Affiliation(s)
- Ji-Seon Ahn
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eun-Jung Ann
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Mi-Yeon Kim
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ji-Hye Yoon
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hye-Jin Lee
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eun-Hye Jo
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Keesook Lee
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ji Shin Lee
- Department of Pathology, Chonnam National University Medical School and Research Institute of Medical Sciences, Gwangju 61469, Republic of Korea
| | - Hee-Sae Park
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
34
|
STAT3 as a promising chemoresistance biomarker associated with the CD44 +/high /CD24 -/low /ALDH + BCSCs-like subset of the triple-negative breast cancer (TNBC) cell line. Exp Cell Res 2018; 363:283-290. [DOI: 10.1016/j.yexcr.2018.01.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/02/2018] [Accepted: 01/12/2018] [Indexed: 12/31/2022]
|
35
|
Fedele M, Cerchia L, Chiappetta G. The Epithelial-to-Mesenchymal Transition in Breast Cancer: Focus on Basal-Like Carcinomas. Cancers (Basel) 2017; 9:cancers9100134. [PMID: 28974015 PMCID: PMC5664073 DOI: 10.3390/cancers9100134] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/13/2017] [Accepted: 09/28/2017] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is a heterogeneous disease that is characterized by a high grade of cell plasticity arising from the contribution of a diverse range of factors. When combined, these factors allow a cancer cell to transition from an epithelial to a mesenchymal state through a process of dedifferentiation that confers stem-like features, including chemoresistance, as well as the capacity to migrate and invade. Understanding the complex events that lead to the acquisition of a mesenchymal phenotype will therefore help to design new therapies against metastatic breast cancer. Here, we recapitulate the main endogenous molecular signals involved in this process, and their cross-talk with paracrine factors. These signals and cross-talk include the extracellular matrix; the secretome of cancer-associated fibroblasts, macrophages, cancer stem cells, and cancer cells; and exosomes with their cargo of miRNAs. Finally, we highlight some of the more promising therapeutic perspectives based on counteracting the epithelial-to-mesenchymal transition in breast cancer cells.
Collapse
Affiliation(s)
- Monica Fedele
- CNR-Institute of Experimental Endocrinology and Oncology, 80131 Naples, Italy.
| | - Laura Cerchia
- CNR-Institute of Experimental Endocrinology and Oncology, 80131 Naples, Italy.
| | - Gennaro Chiappetta
- Dipartimento di Ricerca Traslazionale a Supporto dei Percorsi Oncologici, S.C. Genomica Funzionale, Istituto Nazionale Tumori-IRCCS-Fondazione G Pascale, 80131 Naples, Italy.
| |
Collapse
|
36
|
Bose B, Sen U, Shenoy P S. Breast Cancer Stem Cell Therapeutics, Multiple Strategies Versus Using Engineered Mesenchymal Stem Cells With Notch Inhibitory Properties: Possibilities and Perspectives. J Cell Biochem 2017; 119:141-149. [PMID: 28590064 DOI: 10.1002/jcb.26196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 01/07/2023]
Abstract
Relapse cases of cancers are more vigorous and difficult to control due to the preponderance of cancer stem cells (CSCs). Such CSCs that had been otherwise dormant during the first incidence of cancer gradually appear as radiochemoresistant cancer cells. Hence, cancer therapeutics aimed at CSCs would be an effective strategy for mitigating the cancers during relapse. Alternatively, CSC therapy can also be proposed as an adjuvant therapy, along-with the conventional therapies. As regenerative stem cells (RSCs) are known for their trophic effects, anti-tumorogenicity, and better migration toward an injury site, this review aims to address the use of adult stem cells such as dental pulp derived; cord blood derived pure populations of regenerative stem cells for targeting CSCs. Indeed, pro-tumorogenicity of RSCs is of concern and hence has also been dealt with in relation to breast CSC therapeutics. Furthermore, as notch signaling pathways are upregulated in breast cancers, and anti-notch antibody based and sh-RNA based therapies are already in the market, this review focuses the possibilities of engineering RSCs to express notch inhibitory proteins for breast CSC therapeutics. Also, we have drawn a comparison among various possibilities of breast CSC therapeutics, about, notch1 inhibition. J. Cell. Biochem. 119: 141-149, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bipasha Bose
- Stem Cells and Regenerative Medicine Center, Yenepoya Research Center, Yenepoya University, University Road, Mangalore 575018, Karnataka, India
| | - Utsav Sen
- Stem Cells and Regenerative Medicine Center, Yenepoya Research Center, Yenepoya University, University Road, Mangalore 575018, Karnataka, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Center, Yenepoya Research Center, Yenepoya University, University Road, Mangalore 575018, Karnataka, India
| |
Collapse
|
37
|
Shui Y, Yu X, Duan R, Bao Q, Wu J, Yuan H, Ma C. miR-130b-3p inhibits cell invasion and migration by targeting the Notch ligand Delta-like 1 in breast carcinoma. Gene 2017; 609:80-87. [PMID: 28163094 DOI: 10.1016/j.gene.2017.01.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/30/2017] [Indexed: 02/08/2023]
Abstract
Breast carcinoma is the most common malignancy in women, and the incidence rate has increased dramatically in recent years. Metastasis is responsible for most advanced breast cancer mortality, but the underlying mechanisms remain poorly understood despite extensive research. Recently, short non-coding RNA molecules, including miRNAs, which mediate changes in signalling pathways, have emerged as metastatic regulators of the breast carcinoma. Previous reports have suggested that miR-130b-3p has both oncogenic and tumour suppressor functions in a cancer type-dependent manner. However, the roles and underlying molecular mechanisms of miR-130b-3p in the development of metastasis in breast carcinoma remain unclear. Here, we reported for the first time that miR-130b-3p was differentially expressed in early-stage non-invasive MCF-7 human breast carcinoma cells and aggressive late-stage MDA-MB-231 cells. In gain-of-function and loss-of-function studies, we demonstrated that miR-130b-3p could inhibit breast carcinoma cell invasion and migration by directly targeting the Notch ligand Delta-like 1 (DLL1). Our data also indicated that MMP-9, MMP-13, and VEGF were regulated by miR-130b-3p and may be involved in the inhibition of cell invasion and migration in breast carcinoma. Collectively, our findings reveal a new regulatory mechanism of miR-130b-3p and suggest that miR-130b-3p may be a potential target against human breast cancer metastasis.
Collapse
Affiliation(s)
- Yifang Shui
- Department of Developmental Genetics, Nanjing Medical University, Longmian Road101, Nanjing 211166, P.R. China
| | - Xiaojing Yu
- Department of Developmental Genetics, Nanjing Medical University, Longmian Road101, Nanjing 211166, P.R. China
| | - Rui Duan
- Department of Developmental Genetics, Nanjing Medical University, Longmian Road101, Nanjing 211166, P.R. China
| | - Qianyi Bao
- Department of Developmental Genetics, Nanjing Medical University, Longmian Road101, Nanjing 211166, P.R. China
| | - Jiahui Wu
- Department of Developmental Genetics, Nanjing Medical University, Longmian Road101, Nanjing 211166, P.R. China
| | - Hongyan Yuan
- Department of Oncology and Lombardi Comprehensive Cancer Center, Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| | - Changyan Ma
- Department of Developmental Genetics, Nanjing Medical University, Longmian Road101, Nanjing 211166, P.R. China.
| |
Collapse
|
38
|
Zhou YF, Sun Q, Zhang YJ, Wang GM, He B, Qi T, Zhou Y, Li XW, Li S, He L. Targeted inhibition of Notch1 gene enhances the killing effects of paclitaxel on triple negative breast cancer cells. ASIAN PAC J TROP MED 2017; 10:179-183. [PMID: 28237486 DOI: 10.1016/j.apjtm.2017.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/16/2016] [Accepted: 01/15/2017] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE To study the influence of targeted inhibition of Notch1 gene on the killing effects of paclitaxel on triple negative breast cancer cells. METHODS The triple negative [estrogen receptor (ER)/progesterone receptor (PR)/human epidermal growth factor receptor 2 (Her2)] breast cancer cell line MDA-MB-231 and ER/PR/HER-2-positive breast cancer cell line MCF-7 were cultured, transfected with Notch1-siRNA-overexpression plasmid and blank plasmid, and treated with different concentrations of paclitaxel, and then the cell proliferation activity and apoptosis rate as well as the mRNA expression of Caspase-3, Caspase-9 and Bcl-2 were determined. RESULTS Paclitaxel could decrease the MDA-MB-231 and MCF-7 cell proliferation activity as well as Bcl-2 mRNA expression, and increase MDA-MB-231 and MCF-7 cell apoptosis rate as well as Caspase-3 and Caspase-9 mRNA expression in dose-dependent manners; with the same dose of paclitaxel treatment, the inhibitory effects on MDA-MB-231 cell proliferation activity and Bcl-2 mRNA expression as well as the promoting effects on MDA-MB-231 cell apoptosis and mRNA expression of Caspase-3 and Caspase-9 were weaker than those on MCF-7 cell; after 0.5 μM paclitaxel combined with Notch1-siRNA treatment, MDA-MB-231 cell proliferation activity and Bcl-2 mRNA expression were significantly lower than those after 0.5 μM paclitaxel combined with control plasmid treatment while cell apoptosis rate and mRNA expression of Caspase-3 and Caspase-9 were higher than those after 0.5 μM paclitaxel combined with control plasmid treatment. CONCLUSIONS Targeted inhibition of Notch1 gene may enhance the killing effects of paclitaxel on triple negative breast cancer cells by up-regulating the expression of Caspase-3 and Caspase-9 and inhibiting the expression of Bcl-2.
Collapse
Affiliation(s)
- Yu-Fu Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, Anhui, China.
| | - Qian Sun
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, Anhui, China
| | - Ya-Jun Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, Anhui, China
| | - Geng-Ming Wang
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, Anhui, China
| | - Bin He
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, Anhui, China
| | - Tao Qi
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, Anhui, China
| | - Yan Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, Anhui, China
| | - Xing-Wang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Sheng Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
39
|
Brock EJ, Ji K, Reiners JJ, Mattingly RR. How to Target Activated Ras Proteins: Direct Inhibition vs. Induced Mislocalization. Mini Rev Med Chem 2016; 16:358-69. [PMID: 26423696 DOI: 10.2174/1389557515666151001154002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 09/03/2015] [Accepted: 09/18/2015] [Indexed: 12/13/2022]
Abstract
Oncogenic Ras proteins are a driving force in a significant set of human cancers and wildtype, unmutated Ras proteins likely contribute to the malignant phenotype of many more. The overall challenge of targeting activated Ras proteins has great promise to treat cancer, but this goal has yet to be achieved. Significant efforts and resources have been committed to inhibiting Ras, but these energies have so far made little impact in the clinic. Direct attempts to target activated Ras proteins have faced many obstacles, including the fundamental nature of the gain-of-function oncogenic activity being produced by a loss-of-function at the biochemical level. Nevertheless, there has been very promising recent pre-clinical progress. The major strategy that has so far reached the clinic aimed to inhibit activated Ras indirectly through blocking its post-translational modification and inducing its mislocalization. While these efforts to indirectly target Ras through inhibition of farnesyl transferase (FTase) were rationally designed, this strategy suffered from insufficient attention to the distinctions between the isoforms of Ras. This led to subsequent failures in large-scale clinical trials targeting K-Ras driven lung, colon, and pancreatic cancers. Despite these setbacks, efforts to indirectly target activated Ras through inducing its mislocalization have persisted. It is plausible that FTase inhibitors may still have some utility in the clinic, perhaps in combination with statins or other agents. Alternative approaches for inducing mislocalization of Ras through disruption of its palmitoylation cycle or interaction with chaperone proteins are in early stages of development.
Collapse
Affiliation(s)
| | | | | | - Raymond R Mattingly
- Department of Pharmacology, Wayne State University School of Medicine, 540 East Canfield Ave, Detroit MI, USA.
| |
Collapse
|
40
|
Liu J, Shen JX, Wen XF, Guo YX, Zhang GJ. Targeting Notch degradation system provides promise for breast cancer therapeutics. Crit Rev Oncol Hematol 2016; 104:21-29. [PMID: 27263934 DOI: 10.1016/j.critrevonc.2016.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 04/18/2016] [Accepted: 05/19/2016] [Indexed: 02/05/2023] Open
Abstract
Notch receptor signaling pathways play an important role, not only in normal breast development but also in breast cancer development and progression. As a group of ligand-induced proteins, different subtypes of mammalian Notch (Notch1-4) are sensitive to subtle changes in protein levels. Thus, a clear understanding of mechanisms of Notch protein turnover is essential for understanding normal and pathological mechanisms of Notch functions. It has been suggested that there is a close relationship between the carcinogenesis and the dysregulation of Notch degradation. However, this relationship remains mostly undefined in the context of breast cancer, as protein degradation is mediated by numerous signaling pathways as well as certain molecule modulators (activators/inhibitors). In this review, we summarize the published data regarding the regulation of Notch family member degradation in breast cancer, while emphasizing areas that are likely to provide new therapeutic modalities for mechanism-based anti-cancer drugs.
Collapse
Affiliation(s)
- Jing Liu
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Jia-Xin Shen
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Xiao-Fen Wen
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Yu-Xian Guo
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Guo-Jun Zhang
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| |
Collapse
|
41
|
Geng J, Liu Y, Jin Y, Tai J, Zhang J, Xiao X, Chu P, Yu Y, Wang SC, Lu J, Han S, Shi J, Guo Y, Ni X. MicroRNA-365a-3p promotes tumor growth and metastasis in laryngeal squamous cell carcinoma. Oncol Rep 2016; 35:2017-26. [PMID: 26883008 DOI: 10.3892/or.2016.4617] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/10/2015] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs) are increasingly recognized as oncogenes or tumor suppressors in laryngeal squamous cell carcinoma (LSCC). In this study, we analyzed the roles of miR-365a-3p, miR-143-5p, and miR-494-3p in LSCC using Annexin V/propidium iodide double staining and flow cyto-metry, along with a Transwell migration and invasion assay. The results showed that miR-365a-3p inhibitor significantly facilitated cell apoptosis and suppressed cell cycle progression, migration, and invasion in Hep-2 cells. However, miR-143-5p and miR-494-3p had no such influences. We then investigated the role of miR-365a-3p in LSCC in vivo and found that miR-365a-3p inhibitor suppressed LSCC xenograft tumor growth and metastasis in xenograft mouse models. Moreover, miR-365a-3p inhibitor significantly decreased the expression of p-AKT (Ser473), which indicated that miR-365a-3p can mediate PI3K/AKT signaling pathway transduction via p-AKT (Ser473) in LSCC. The data suggest that miR-365a-3p may act as an oncomiR and may promote growth and metastasis in LSCC via the PI3K/AKT signaling pathway, and thus miR‑365a-3p may be a potential therapeutic target for treatment of LSCC.
Collapse
Affiliation(s)
- Jiangqiao Geng
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Yuanhu Liu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jun Tai
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Xiao Xiao
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Ping Chu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Yongbo Yu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Sheng Cai Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jie Lu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Shujing Han
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jin Shi
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| |
Collapse
|
42
|
Jung YC, Han S, Hua L, Ahn YH, Cho H, Lee CJ, Lee H, Cho YY, Ryu JH, Jeon R, Kim WY. Kazinol-E is a specific inhibitor of ERK that suppresses the enrichment of a breast cancer stem-like cell population. Biochem Biophys Res Commun 2016; 470:294-299. [PMID: 26774343 DOI: 10.1016/j.bbrc.2016.01.066] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 01/10/2016] [Indexed: 01/16/2023]
Abstract
Growing evidence shows that cancer stem-like cells (CSLCs) contribute to breast cancer recurrence and to its resistance to conventional therapies. The extracellular signal-regulated kinase (ERK) signaling pathway is a major determinant in the control of diverse cellular processes, including the maintenance of CSLCs. In this study, we found that Kazinol-E, an antioxidant flavan from Broussonetia kazinoki, decreased the CSLC population of a breast cancer cell line, MCF7. The CSLC population, characterized by CD44 high/CD24 low expression or by high Aldehyde dehydrogenase 1 activity, was decreased by a concentration of Kazinol-E that did not affect the growth of bulk-cultured MCF7 cells. Kazinol-E did not decrease EGF-induced ERK phosphorylation in CSLCs, but did block the phosphorylation of an ERK substrate, p90RSK2, at Thr359/Ser363. We further demonstrated that EGF-induced ERK activity was blocked by Kazinol-E in a wild-type K-Ras-expressing non-small cell lung cancer cell line H226B. An in vitro kinase assay with purified ERK1 and p90RSK2 as its substrate demonstrated a direct inhibition of ERK activity by Kazinol E. Additionally, a the molecular docking study provided putative binding modes of Kazinol-E into the ATP binding pocket of ERK1 Collectively, these results suggest that Kazinol-E is a direct inhibitor of ERK1, and more studies are warranted to develop this reagent for therapeutic breast CSLC targeting.
Collapse
Affiliation(s)
- Yu-Chae Jung
- The Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Seula Han
- The Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Li Hua
- The Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Yeon-Hwa Ahn
- The Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Hyewon Cho
- The Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Cheol-Jung Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hani Lee
- The Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Jae-Ha Ryu
- The Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Raok Jeon
- The Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Woo-Young Kim
- The Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea.
| |
Collapse
|
43
|
Zheng MW, Zhang CH, Chen K, Huang M, Li YP, Lin WT, Zhang RJ, Zhong L, Xiang R, Li LL, Liu XY, Wei YQ, Yang SY. Preclinical Evaluation of a Novel Orally Available SRC/Raf/VEGFR2 Inhibitor, SKLB646, in the Treatment of Triple-Negative Breast Cancer. Mol Cancer Ther 2015; 15:366-78. [PMID: 26721945 DOI: 10.1158/1535-7163.mct-15-0501] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/20/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Ming-Wu Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Chun-Hui Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Kai Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Mei Huang
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Ya-Ping Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Wan-Ting Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Rong-Jie Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Lei Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Rong Xiang
- Department of Clinical Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Lin-Li Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Xin-Yu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Yu-Quan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Sheng-Yong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China.
| |
Collapse
|
44
|
Notch signaling: an emerging therapeutic target for cancer treatment. Cancer Lett 2015; 369:20-7. [PMID: 26341688 DOI: 10.1016/j.canlet.2015.07.048] [Citation(s) in RCA: 298] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/28/2015] [Accepted: 07/31/2015] [Indexed: 12/14/2022]
Abstract
The Notch pathway is involved in cell proliferation, differentiation and survival. The Notch signaling pathway is one of the most commonly activated signaling pathways in cancer. Alterations include activating mutations and amplification of the Notch pathway, which play key roles in the progression of cancer. Accumulating evidence suggests that the pharmacological inhibition of this pathway can overcome chemoresistance. Efforts have been taken to develop Notch inhibitors as a single agent or in combination with clinically used chemotherapeutics to treat cancer. Some Notch inhibitors have been demonstrated to have therapeutic efficacy in preclinical studies. This review summarizes the recent studies and clinical evaluations of the Notch inhibitors in cancer.
Collapse
|
45
|
Yuan X, Zhang M, Wu H, Xu H, Han N, Chu Q, Yu S, Chen Y, Wu K. Expression of Notch1 Correlates with Breast Cancer Progression and Prognosis. PLoS One 2015; 10:e0131689. [PMID: 26121683 PMCID: PMC4488260 DOI: 10.1371/journal.pone.0131689] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/05/2015] [Indexed: 02/07/2023] Open
Abstract
Various studies have evaluated the significance of Notch1 expression in breast cancer, but the results have ever been disputed. By using 21 studies involving 3867 patients, this meta-analysis revealed that the expression of Notch1 was significantly higher in breast cancer than in normal tissues (OR=7.21; 95%CI, 4.7-11.07) and that higher Notch1 expression was associated with transition from ductal carcinoma in situ (DCIS) to invasive cancer (OR=3.75; 95% CI, 1.8-7.78). Higher Notch1 activity was observed in the basal subtype of breast cancer (OR=2.53; 95% CI, 1.18-5.43). Moreover, patients with Notch1 overexpression exhibited significantly worse overall and recurrence-free survival. Our meta-analysis suggests that Notch inhibitors may be useful in blocking the early progression of DCIS and that the outcomes of clinical trials for Notch1-targeting therapeutics could be improved by the molecular stratification of breast cancer patients.
Collapse
Affiliation(s)
- Xun Yuan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Mingsheng Zhang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hua Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hanxiao Xu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Na Han
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Shiying Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yuan Chen
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- * E-mail:
| |
Collapse
|