1
|
Glass K, Fines C, Coulter P, Jena L, McCarthy HO, Buckley N. Development and Characterization of a Peptide-Bisphosphonate Nanoparticle for the Treatment of Breast Cancer. Mol Pharm 2024; 21:4970-4982. [PMID: 39196792 PMCID: PMC11462496 DOI: 10.1021/acs.molpharmaceut.4c00299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 08/30/2024]
Abstract
In women, breast cancer (BC) is the most common cancer, and despite advancements in diagnosis and treatment, 20-30% of early stage BC patients develop metastatic disease. Metastatic BC is deemed an incurable disease, which accounts for 90% of BC related deaths, with only 26% of metastatic patients reaching a 5 year survival rate. Therefore, there is an unmet need for the prevention or treatment of metastasis in early stage breast cancer patients. Bisphosphonates (BPs) are potent inhibitors of bone resorption and are extensively used for the prevention of osteoporosis and other skeletal disorders, as well as for the treatment of secondary bone cancer in BC patients. Furthermore, the direct anticancer activity of BPs has been established in primary tumor models. However, these studies were limited by the need for dosages far above the clinical range to overcome BPs' high affinity for bones and poor accumulation in the tumor itself, which leads to toxicity, including osteonecrosis of the jaw. To decrease BP dosage, increase bioavailability, and direct anticancer activity, we used the RALA (R-) peptide delivery system to form highly stable NPs with the nitrogen containing BP, risedronate (R-RIS). In vitro studies showed that, in comparison to RIS, R-RIS nanoparticles increased cytotoxicity and reduced metastatic features such as proliferation, migration, invasion, and adhesion of metastatic BC cells to bones. Furthermore, in an in vivo model, R-RIS had increased tumor accumulation while still maintaining similar bone accumulation to RIS alone. This increase in tumor accumulation corresponded with decreased tumor volume and lungs metastasis. R-RIS has great potential to be used in combination with standard of care chemotherapy for the treatment of primary BC and its metastasis while still having its bone resorption inhibiting properties.
Collapse
Affiliation(s)
- Kimberley Glass
- School of Pharmacy, Queen’s
University Belfast, 97 Lisburn Road, BT9 7BL Northern Ireland, U.K.
| | - Cory Fines
- School of Pharmacy, Queen’s
University Belfast, 97 Lisburn Road, BT9 7BL Northern Ireland, U.K.
| | - Paula Coulter
- School of Pharmacy, Queen’s
University Belfast, 97 Lisburn Road, BT9 7BL Northern Ireland, U.K.
| | - Lynn Jena
- School of Pharmacy, Queen’s
University Belfast, 97 Lisburn Road, BT9 7BL Northern Ireland, U.K.
| | - Helen O. McCarthy
- School of Pharmacy, Queen’s
University Belfast, 97 Lisburn Road, BT9 7BL Northern Ireland, U.K.
| | - Niamh Buckley
- School of Pharmacy, Queen’s
University Belfast, 97 Lisburn Road, BT9 7BL Northern Ireland, U.K.
| |
Collapse
|
2
|
Zin I, China A, Khan K, Nag JK, Vasu K, Deshpande GM, Ghosh PK, Khan D, Ramachandiran I, Ganguly S, Tamagno I, Willard B, Gogonea V, Fox PL. AKT-dependent nuclear localization of EPRS1 activates PARP1 in breast cancer cells. Proc Natl Acad Sci U S A 2024; 121:e2303642121. [PMID: 39012819 PMCID: PMC11287164 DOI: 10.1073/pnas.2303642121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/05/2024] [Indexed: 07/18/2024] Open
Abstract
Glutamyl-prolyl-tRNA synthetase (EPRS1) is a bifunctional aminoacyl-tRNA-synthetase (aaRS) essential for decoding the genetic code. EPRS1 resides, with seven other aaRSs and three noncatalytic proteins, in the cytoplasmic multi-tRNA synthetase complex (MSC). Multiple MSC-resident aaRSs, including EPRS1, exhibit stimulus-dependent release from the MSC to perform noncanonical activities distinct from their primary function in protein synthesis. Here, we show EPRS1 is present in both cytoplasm and nucleus of breast cancer cells with constitutively low phosphatase and tensin homolog (PTEN) expression. EPRS1 is primarily cytosolic in PTEN-expressing cells, but chemical or genetic inhibition of PTEN, or chemical or stress-mediated activation of its target, AKT, induces EPRS1 nuclear localization. Likewise, preferential nuclear localization of EPRS1 was observed in invasive ductal carcinoma that were also P-Ser473-AKT+. EPRS1 nuclear transport requires a nuclear localization signal (NLS) within the linker region that joins the catalytic glutamyl-tRNA synthetase and prolyl-tRNA synthetase domains. Nuclear EPRS1 interacts with poly(ADP-ribose) polymerase 1 (PARP1), a DNA-damage sensor that directs poly(ADP-ribosyl)ation (PARylation) of proteins. EPRS1 is a critical regulator of PARP1 activity as shown by markedly reduced ADP-ribosylation in EPRS1 knockdown cells. Moreover, EPRS1 and PARP1 knockdown comparably alter the expression of multiple tumor-related genes, inhibit DNA-damage repair, reduce tumor cell survival, and diminish tumor sphere formation by breast cancer cells. EPRS1-mediated regulation of PARP1 activity provides a mechanistic link between PTEN loss in breast cancer cells, PARP1 activation, and cell survival and tumor growth. Targeting the noncanonical activity of EPRS1, without inhibiting canonical tRNA ligase activity, provides a therapeutic approach potentially supplementing existing PARP1 inhibitors.
Collapse
Affiliation(s)
- Isaac Zin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
- Department of Chemistry, Cleveland State University, Cleveland, OH44115
| | - Arnab China
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Krishnendu Khan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
- Department of Life Sciences, School of Science, Gandhi Institute of Technology and Management, Bengaluru562163, India
| | - Jeetendra K. Nag
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Kommireddy Vasu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | | | - Prabar K. Ghosh
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Debjit Khan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Iyappan Ramachandiran
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Shinjini Ganguly
- Translational Hematology and Oncology Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Ilaria Tamagno
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Belinda Willard
- Proteomics and Metabolomics Core, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH44195
| | - Valentin Gogonea
- Department of Chemistry, Cleveland State University, Cleveland, OH44115
| | - Paul L. Fox
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
- Department of Chemistry, Cleveland State University, Cleveland, OH44115
| |
Collapse
|
3
|
Du SG, Zhang HM, Ji YX, Tian YL, Wang D, Zhu K, Zhang QG, Liu SP. Polyphyllin VII Promotes Apoptosis in Breast Cancer by Inhibiting MAPK/ERK Signaling Pathway through Downregulation of SOS1. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:885-904. [PMID: 38716619 DOI: 10.1142/s0192415x24500368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Polyphyllin VII is a biologically active herbal monomer extracted from the traditional Chinese herbal medicine Chonglou. Many studies have demonstrated the anticancer activity of polyphyllin VII against various types of cancers, such as colon, liver, and lung cancer, but its effect on breast cancer has not been elucidated. In this study, we demonstrate that polyphyllin VII inhibited proliferation, increased production of intracellular reactive oxygen species, and decreased mitochondrial membrane potential in breast cancer cells. Notably, polyphyllin VII also induced apoptosis via the mitochondrial pathway. Transcriptome sequencing was used to analyze the targets of PPVII in regulating breast cancer cells. Mechanistic studies showed that polyphyllin VII downregulated Son of Sevenless1 (SOS1) and inhibited the MAPK/ERK pathway. Furthermore, PPVII exerted strong antitumor effects in vivo in nude mice injected with breast cancer cells. Our results suggest that PPVII may promote apoptosis through regulating the SOS1/MAPK/ERK pathway, making it a possible candidate target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shu-Guang Du
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
- Laboratory Department, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, P. R. China
| | - Hua-Min Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, P. R. China
| | - Yun-Xia Ji
- Laboratory Department, Zhangjiakou First Hospital, Zhangjiakou 075041, P. R. China
| | - Yu-Lin Tian
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| | - Dan Wang
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| | - Kun Zhu
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| | - Qing-Gao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| | - Shuang-Ping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| |
Collapse
|
4
|
Szablewski L. Changes in Cells Associated with Insulin Resistance. Int J Mol Sci 2024; 25:2397. [PMID: 38397072 PMCID: PMC10889819 DOI: 10.3390/ijms25042397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Insulin is a polypeptide hormone synthesized and secreted by pancreatic β-cells. It plays an important role as a metabolic hormone. Insulin influences the metabolism of glucose, regulating plasma glucose levels and stimulating glucose storage in organs such as the liver, muscles and adipose tissue. It is involved in fat metabolism, increasing the storage of triglycerides and decreasing lipolysis. Ketone body metabolism also depends on insulin action, as insulin reduces ketone body concentrations and influences protein metabolism. It increases nitrogen retention, facilitates the transport of amino acids into cells and increases the synthesis of proteins. Insulin also inhibits protein breakdown and is involved in cellular growth and proliferation. On the other hand, defects in the intracellular signaling pathways of insulin may cause several disturbances in human metabolism, resulting in several chronic diseases. Insulin resistance, also known as impaired insulin sensitivity, is due to the decreased reaction of insulin signaling for glucose levels, seen when glucose use in response to an adequate concentration of insulin is impaired. Insulin resistance may cause, for example, increased plasma insulin levels. That state, called hyperinsulinemia, impairs metabolic processes and is observed in patients with type 2 diabetes mellitus and obesity. Hyperinsulinemia may increase the risk of initiation, progression and metastasis of several cancers and may cause poor cancer outcomes. Insulin resistance is a health problem worldwide; therefore, mechanisms of insulin resistance, causes and types of insulin resistance and strategies against insulin resistance are described in this review. Attention is also paid to factors that are associated with the development of insulin resistance, the main and characteristic symptoms of particular syndromes, plus other aspects of severe insulin resistance. This review mainly focuses on the description and analysis of changes in cells due to insulin resistance.
Collapse
Affiliation(s)
- Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
5
|
Keesari PR, Jain A, Ganampet NR, Subhasri GSD, Edusa S, Muslehuddin Z, Theik NWY, Palisetti S, Salibindla D, Manaktala PS, Desai R. Association between prediabetes and breast cancer: a comprehensive meta-analysis. Breast Cancer Res Treat 2024; 204:1-13. [PMID: 38060076 DOI: 10.1007/s10549-023-07181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Breast cancer accounts for up to 30% of cancer cases in women in the US. Diabetes mellitus has been recognized as a risk factor for breast cancer. Some studies have suggested that prediabetes may also be associated with breast cancer whereas other studies have shown no or an inverse association; thus, we conducted a meta-analysis to assess the risk of breast cancer in prediabetes. METHODS We searched PubMed/Medline, EMBASE, Google Scholar, and Scopus to identify studies that reported breast cancer risks in patients having prediabetes compared to normoglycemic patients. Binary random-effects model was used to calculate a pooled odds ratio (OR) with 95% confidence intervals. I2 statistics were used to assess heterogeneity. Leave-one-out sensitivity analysis and subgroup analyses were performed. RESULTS We analyzed 7 studies with 24,586 prediabetic and 224,314 normoglycemic individuals (783 and 5739 breast cancer cases, respectively). Unadjusted odds ratio (OR) for breast cancer was 1.45 (95% CI = 1.14, 1.83); adjusted OR was 1.19 (95% CI = 1.07, 1.34) in prediabetes. Subgroup analysis revealed a higher breast cancer risk in individuals aged less than 60 years (OR = 1.86, 95% CI = 1.39, 2.49) than in those aged 60 years or more (OR = 1.07, 95% CI = 0.97, 1.18). Subgroup analysis by median follow-up length indicated a higher risk of breast cancer for follow-ups of less than or equal to 2 years (OR = 2.34, 95% CI = 1.85, 2.95) than in those of over 10 years (OR = 1.1, 95% CI = 0.99, 1.23) and 6 to 10 years (OR = 1.03, 95% CI = 0.88, 1.21). CONCLUSIONS In conclusion, individuals with prediabetes have higher risk of developing breast cancer than those with normoglycemia, especially younger prediabetes patients. These individuals may benefit from early identification, monitoring, and interventions to reverse prediabetes.
Collapse
Affiliation(s)
- Praneeth Reddy Keesari
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Akhil Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | | | | | - Samuel Edusa
- Department of Internal Medicine, Piedmont Athens Regional, Athens, GA, USA
| | - Zainab Muslehuddin
- Department of Internal Medicine, Detroit Medical Center, Wayne State University-Sinai Grace Hospital, Detroit, MI, USA
| | | | - Spandana Palisetti
- Department of Medicine, Jawaharlal Nehru Medical College, Belgaum, Karnataka, India
| | | | | | - Rupak Desai
- Independent Researcher, Outcomes Research, Atlanta, GA, USA
| |
Collapse
|
6
|
Hayama S, Nakamura R, Ishige T, Sangai T, Sakakibara M, Fujimoto H, Ishigami E, Masuda T, Nakagawa A, Teranaka R, Ota S, Itoga S, Yamamoto N, Nagashima T, Otsuka M. The impact of PIK3CA mutations and PTEN expression on the effect of neoadjuvant therapy for postmenopausal luminal breast cancer patients. BMC Cancer 2023; 23:384. [PMID: 37106324 PMCID: PMC10134571 DOI: 10.1186/s12885-023-10853-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND There is pressing needs to find the biomarker in the selection of neoadjuvant therapy in postmenopausal luminal breast cancer patients. We examined the hypothesis that PIK3CA mutations and low phosphatase and tensin homolog (PTEN) expression affect the response to neoadjuvant therapy and prognosis in postmenopausal luminal breast cancer patients. METHODS Postmenopausal patients with estrogen receptor-positive, human epidermal growth factor receptor 2-negative breast cancer, up to stage II, who underwent neoadjuvant chemotherapy (NAC; n = 60) or neoadjuvant endocrine therapy (NAE; n = 55) were selected. PIK3CA exon 9 and exon 20 mutations were screened by high resolution melting analysis and confirmed by Sanger sequence. PTEN expression was evaluated by immunohistochemistry. The relationships among PIK3CA mutations, PTEN expression, clinicopathological features, the pathological effect of neoadjuvant therapy, recurrence-free survival (RFS) and overall survival were analyzed. RESULTS Among 115 patients, PIK3CA mutations and low PTEN expression before treatment were detected in 35 patients (30.4%) and in 28 patients (24.3%), respectively. In the NAC group, tumor with PIK3CA mutations showed significantly poorer response than tumor with PIK3CA wild-type (p = 0.03). On the other hand, in the NAE group, there was no significant difference in pathological therapeutic effect between tumor with PIK3CA mutations and tumor with PIK3CA wild-type (p = 0.54). In the NAC group, the log-rank test showed no difference in RFS between patients with PIK3CA mutations and PIK3CA wild-type (p = 0.43), but patients with low PTEN expression showed significantly worse RFS compared to patients with high PTEN expression (5 year RFS 0.64 vs. 0.87, p = 0.01). In the Cox proportional hazards model for RFS, PTEN expression, progesterone receptor, and pathological therapeutic effect were predictive factors for time to recurrence (All p < 0.05). CONCLUSIONS PIK3CA mutations are associated with resistance to NAC but do not affect the response to NAE. Low PTEN expression does not affect response to either NAC or NAE but correlates with shorter RFS in patients who received NAC. These biomarkers will be further evaluated for clinical use to treat postmenopausal luminal breast cancer patients.
Collapse
Affiliation(s)
- Shouko Hayama
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
- Department of Breast Surgery, Chiba Cancer Center, 666-2 Nitona, Chuo-Ku, Chiba-Shi, Chiba, 260-8717, Japan
| | - Rikiya Nakamura
- Department of Breast Surgery, Chiba Cancer Center, 666-2 Nitona, Chuo-Ku, Chiba-Shi, Chiba, 260-8717, Japan
| | - Takayuki Ishige
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Takafumi Sangai
- Department of Breast and Thyroid Surgery, Kitasato University School of Medicine, 1-15-1 Kitazato Minami-Ku, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Masahiro Sakakibara
- Departments of Breast Surgery, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura, Chiba, 285-8741, Japan
| | - Hiroshi Fujimoto
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Emi Ishigami
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Takahito Masuda
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Ayako Nakagawa
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Ryotaro Teranaka
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Satoshi Ota
- Department of Pathology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Sakae Itoga
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-6-7 Kazusa-Kamatari, Kisarazu Chiba, 292-0818, Japan
| | - Naohito Yamamoto
- Department of Breast Surgery, Chiba Cancer Center, 666-2 Nitona, Chuo-Ku, Chiba-Shi, Chiba, 260-8717, Japan
| | - Takeshi Nagashima
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Masayuki Otsuka
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| |
Collapse
|
7
|
Kerdjoudj M, De La Torre RA, Arnouk H. Characterization of DJ-1, PTEN, and p-Akt as Prognostic Biomarkers in the Progression of Oral Squamous Cell Carcinoma. Cureus 2023; 15:e34436. [PMID: 36874678 PMCID: PMC9979586 DOI: 10.7759/cureus.34436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 02/01/2023] Open
Abstract
Objective Oral cancer has a five-year survival rate of 68%, and the methods used to assess it still rely heavily on morphology. Protein biomarkers can potentially increase the predictive power of histopathological evaluation. This study aims to examine the expression of three closely linked proteins implicated in the pathogenesis of oral squamous cell carcinoma (OSCC); protein deglycase (DJ-1), an oncogene, phosphatase, and tensin homolog (PTEN), a tumor suppressor gene, and phosphorylated protein kinase B (p-Akt), the activated form of a vital serine/threonine kinase, which is involved in the oncogenesis of several human malignancies, throughout the tumor progression steps to establish their potential as prognostic biomarkers. Study design Western blot analysis was carried out using four different cell lines representing the successive steps of OSCC progression, including normal oral keratinocytes, dysplastic oral keratinocytes, locally invasive OSCC, and metastatic OSCC. Results DJ-1 expression was found to be upregulated gradually throughout the successive steps of OSCC progression from normal to dysplastic to locally invasive to metastatic OSCC. PTEN expression showed an overall opposite trend. Interestingly, a significant downregulation of p-Akt was seen in the locally invasive OSCC cells, although it was followed by a significant increase in p-Akt expression in the metastatic OSCC cell line, which is consistent with the role of p-Akt in the motility and migration of cancer cells. Conclusion This study documented trends in expression patterns of three important signaling molecules, DJ-1, PTEN, and p-Akt, in normal, premalignant, and malignant oral keratinocytes. The oncogenic DJ-1 and tumor suppressor PTEN were expressed in a manner consistent with their respective roles in tumorigenesis, while p-Akt only showed a significant upregulation in the metastatic OSCC cells. Overall, all three proteins exhibited unique trends throughout the progressive stages of OSCC tumor progression, thereby adding to their potential utility as prognostic biomarkers for oral cancer patients.
Collapse
Affiliation(s)
- Mourad Kerdjoudj
- Internal Medicine, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, USA
| | - Rey A De La Torre
- Osteopathic Medicine, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, USA
| | - Hilal Arnouk
- Pathology, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, USA
- Pathology, College of Graduate Studies, Midwestern University, Downers Grove, USA
- Pathology, College of Dental Medicine-Illinois, Midwestern University, Downers Grove, USA
- Pathology, Chicago College of Optometry, Midwestern University, Downers Grove, USA
- Molecular Pathology, Precision Medicine Program, Midwestern University, Downers Grove, USA
| |
Collapse
|
8
|
Fazil WFWM, Amanah A, Abduraman MA, Sulaiman SF, Wahab HA, Tan ML. The Effects of Deoxyelephantopin on the Akt/mTOR/P70S6K Signaling Pathway in MCF-7 Breast Carcinoma Cells In Vitro. J Pharmacol Pharmacother 2022. [DOI: 10.1177/0976500x221114003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective To determine the effects of deoxyelephantopin on mTOR and its related target molecules (Akt/mTOR/P70S6K) in the ER-positive breast cancer cell line. Materials and Methods Primary in silico simulations were determined, and the effects of deoxyelephantopin on the phosphorylation of the Akt/mTOR/P70S6K molecules were evaluated using AlphaScreen-based assays and western blot analysis, respectively. Results Based on the estimated FEB and K i values, deoxyelephantopin appeared to have a stronger affinity toward P70S6K as compared with Akt and mTOR. Both deoxyelephantopin and control inhibitors were observed to form hydrogen bonds with the same key residue, Leu175 of the P70S6K molecule. Deoxyelephantopin downregulated the p-P70S6K protein expression significantly from 18 µM ( P < .05) and onward. Based on the AlphaScreen assay, deoxyelephantopin produced a concentration-dependent inhibition on the phosphorylation of P70S6K with an IC50 value of 7.13 µM. Conclusion Deoxyelephantopin induced cell death in MCF-7 cells, possibly via DNA fragmentation, inhibition of the phosphorylation of P70SK6, and downregulation of the relative p-p70S6K protein expression levels.
Collapse
Affiliation(s)
- Wan Failiza Wan Mohamad Fazil
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), National Institutes of Biotechnology Malaysia (NIBM), Pulau Pinang, Malaysia
| | - Azimah Amanah
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), National Institutes of Biotechnology Malaysia (NIBM), Pulau Pinang, Malaysia
| | - Muhammad Asyraf Abduraman
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, SAINS@BERTAM, Kepala Batas, Pulau Pinang, Malaysia
| | - Shaida Fariza Sulaiman
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Pulau Pinang, Malaysia
| | - Habibah Abdul Wahab
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Pulau Pinang, Malaysia
| | - Mei Lan Tan
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), National Institutes of Biotechnology Malaysia (NIBM), Pulau Pinang, Malaysia
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, SAINS@BERTAM, Kepala Batas, Pulau Pinang, Malaysia
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Pulau Pinang, Malaysia
| |
Collapse
|
9
|
The AKT1E17K Allele Promotes Breast Cancer in Mice. Cancers (Basel) 2022; 14:cancers14112645. [PMID: 35681625 PMCID: PMC9179273 DOI: 10.3390/cancers14112645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary The main finding reported in this manuscript is that the gain-of-function mutation AKT1E17K is a bona fide oncogene for mammary epithelium, being able to efficiently initiate breast cancer in mice. On the basis of high-molecular-weight cytokeratins expressed by AKT1E17K-derived tumors supported by additional integrative gene expression analysis these tumors resulted similar to human basal-like cancer, phenotypically and molecularly. These results indicate that the AKTE17K strain may represent an appropriate model of human basal-like breast cancer for the identification of novel therapies specific for this type of tumor. Abstract The gain-of-function mutation in the pleckstrin homology domain of AKT1 (AKT1E17K) occurs in lung and breast cancer. Through the use of human cellular models and of a AKT1E17K transgenic Cre-inducible murine strain (R26-AKT1E17K mice), we have demonstrated that AKT1E17K is a bona fide oncogene for lung epithelial cells. However, the role of AKT1E17K in breast cancer remains to be determined. Here, we report the generation and the characterization of a MMTV-CRE; R26-AKT1E17K mouse strain that expresses the mutant AKT1E17K allele in the mammary epithelium. We observed that AKT1E17K stimulates the development of mammary tumors classified as ductal adenocarcinoma of medium–high grade and presented a variety of proliferative alterations classified as adenosis with low-to-high grade dysplasia in the mammary epithelium. A subsequent immunohistochemical characterization suggested they were PR−/HER2−/ER+, basal-like and CK8−/CK10−/CK5+/CK14+. We also observed that, in parallel with an increased proliferation rate, tumors expressing mutant AKT1E17K presented an activation of the GSK3/cyclin D1 pathway in the mammary epithelium and cluster significantly with the human basal-like tumors. In conclusion, we demonstrate AKT1E17K is a bona fide oncogene that can initiate tumors at high efficiency in murine mammary epithelium in vivo.
Collapse
|
10
|
Sibilano M, Tullio V, Adorno G, Savini I, Gasperi V, Catani MV. Platelet-Derived miR-126-3p Directly Targets AKT2 and Exerts Anti-Tumor Effects in Breast Cancer Cells: Further Insights in Platelet-Cancer Interplay. Int J Mol Sci 2022; 23:ijms23105484. [PMID: 35628294 PMCID: PMC9141257 DOI: 10.3390/ijms23105484] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 02/07/2023] Open
Abstract
Among the surrounding cells influencing tumor biology, platelets are recognized as novel players as they release microvesicles (MVs) that, once delivered to cancer cells, modulate signaling pathways related to cell growth and dissemination. We have previously shown that physiological delivery of platelet MVs enriched in miR-126 exerted anti-tumor effects in different breast cancer (BC) cell lines. Here, we seek further insight by identifying AKT2 kinase as a novel miR-126-3p direct target, as assessed by bioinformatic analysis and validated by luciferase assay. Both ectopic expression and platelet MV-mediated delivery of miR-126-3p downregulated AKT2 expression, thus suppressing proliferating and invading properties, in either triple negative (BT549 cells) or less aggressive Luminal A (MCF-7 cells) BC subtypes. Accordingly, as shown by bioinformatic analysis, both high miR-126 and low AKT2 levels were associated with favorable long-term prognosis in BC patients. Our results, together with the literature data, indicate that miR-126-3p exerts suppressor activity by specifically targeting components of the PIK3/AKT signaling cascade. Therefore, management of platelet-derived MV production and selective delivery of miR-126-3p to tumor cells may represent a useful tool in multimodal therapeutic approaches in BC patients.
Collapse
Affiliation(s)
- Matteo Sibilano
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
| | - Valentina Tullio
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
| | - Gaspare Adorno
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Isabella Savini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
| | - Valeria Gasperi
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
- Correspondence: (V.G.); (M.V.C.); Tel.: +39-06-7259-6465 (V.G.); +39-06-7259-6465 (M.V.C.)
| | - Maria Valeria Catani
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
- Correspondence: (V.G.); (M.V.C.); Tel.: +39-06-7259-6465 (V.G.); +39-06-7259-6465 (M.V.C.)
| |
Collapse
|
11
|
Luque M, Cristóbal I, Sanz-Álvarez M, Santos A, Zazo S, Eroles P, Arpí O, Rovira A, Albanell J, Madoz-Gúrpide J, García-Foncillas J, Rojo F. CIP2A as a Key Regulator for AKT Phosphorylation Has Partial Impact Determining Clinical Outcome in Breast Cancer. J Clin Med 2022; 11:jcm11061610. [PMID: 35329936 PMCID: PMC8955826 DOI: 10.3390/jcm11061610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/03/2022] [Accepted: 03/12/2022] [Indexed: 12/07/2022] Open
Abstract
Together with its reported ability to modulate AKT phosphorylation (p-AKT) status in several tumor types, the oncoprotein CIP2A has been described to induce breast cancer progression and drug resistance. However, the clinical and therapeutic relevance of the CIP2A/AKT interplay in breast cancer remains to be fully clarified. Here, we found high p-AKT levels in 80 out of 220 cases (36.4%), which were associated with negative estrogen receptor expression (p = 0.049) and CIP2A overexpression (p < 0.001). Interestingly, p-AKT determined substantially shorter overall (p = 0.002) and progression-free survival (p = 0.003), and multivariate analyses showed its CIP2A-independent prognostic value. Moreover, its clinical relevance was further confirmed in the triple negative and HER2-positive subgroups after stratifying our series by molecular subtype. Functionally, we confirmed in vitro the role of CIP2A as a regulator of p-AKT levels in breast cancer cell lines, and the importance of the CIP2A/AKT axis was also validated in vivo. Finally, p-AKT also showed a higher predictive value of response to doxorubicin than CIP2A in ex vivo analyses. In conclusion, our findings suggest that CIP2A overexpression is a key contributing event to AKT phosphorylation and highlights the CIP2A/AKT axis as a promising therapeutic target in breast cancer. However, our observations highlight the existence of alternative mechanisms that regulate AKT signaling in a subgroup of breast tumors without altered CIP2A expression that determines its independent value as a marker of poor outcome in this disease.
Collapse
Affiliation(s)
- Melani Luque
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| | - Marta Sanz-Álvarez
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Andrea Santos
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Sandra Zazo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Pilar Eroles
- Institute of Health Research INCLIVA, 46010 Valencia, Spain;
| | - Oriol Arpí
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Juan Madoz-Gúrpide
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Federico Rojo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| |
Collapse
|
12
|
Shaji SK, Drishya G, Sunilkumar D, Suravajhala P, Kumar GB, Nair BG. Systematic understanding of anti-tumor mechanisms of Tamarixetin through network and experimental analyses. Sci Rep 2022; 12:3966. [PMID: 35273218 PMCID: PMC8913656 DOI: 10.1038/s41598-022-07087-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/10/2021] [Indexed: 11/24/2022] Open
Abstract
Tamarixetin, a flavonoid derived from Quercetin, was shown to possess anti-cancer properties in various types of cancer. However, the mechanism of action of this compound is not well understood. Observations from reverse docking and network pharmacology analysis, were validated by cell based studies to analyse the chemotherapeutic potential and elucidate the molecular mechanism of action of Tamarixetin in breast cancer. In silico analysis using reverse docking and PPI analysis clearly indicated that out of 35 proteins targeted by Tamarixetin, the top 3 hub genes, namely, AKT1, ESR1 and HSP90AA1, were upregulated in breast tumor tissues and more importantly showed strong negative correlation to breast cancer patient survival. Furthermore, the KEGG pathway analysis showed enrichment of target proteins of Tamarixetin in 33 pathways which are mainly involved in neoplastic signalling. In vitro cell-based studies demonstrated that Tamarixetin could inhibit cell proliferation, induce ROS and reduce mitochondrial membrane potential, leading to cell death. Tamarixetin induced cell cycle arrest at G2/M phase and inhibited the migration as well as the invasion of breast cancer cells. Taken together, the combination of in silico and in vitro approaches used in the present study clearly provides evidence for the chemotherapeutic potential of Tamarixetin in breast cancer.
Collapse
Affiliation(s)
- Sanu K Shaji
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O, Kollam, Kerala, 690525, India
| | - G Drishya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O, Kollam, Kerala, 690525, India
| | - Damu Sunilkumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O, Kollam, Kerala, 690525, India
| | - Prashanth Suravajhala
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O, Kollam, Kerala, 690525, India
| | - Geetha B Kumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O, Kollam, Kerala, 690525, India.
| | - Bipin G Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O, Kollam, Kerala, 690525, India.
| |
Collapse
|
13
|
Zhou Z, Zhang C, Ma Z, Wang H, Tuo B, Cheng X, Liu X, Li T. Pathophysiological role of ion channels and transporters in HER2-positive breast cancer. Cancer Gene Ther 2022; 29:1097-1104. [PMID: 34997219 DOI: 10.1038/s41417-021-00407-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/21/2021] [Accepted: 11/08/2021] [Indexed: 11/09/2022]
Abstract
The incidence of breast cancer (BC) has been increasing each year, and BC is now the most common malignant tumor in women. Among the numerous BC subtypes, HER2-positive BC can be treated with a variety of strategies based on targeting HER2. Although there has been great progress in the treatment of HER2-positive BC, recurrence, metastasis and drug resistance remain considerable challenges. The dysfunction of ion channels and transporters can affect the development and progression of HER2-positive BC, so these entities are expected to be new therapeutic targets. This review summarizes various ion channels and transporters associated with HER2-positive BC and suggests potential targets for the development of new and effective therapies.
Collapse
Affiliation(s)
- Zhengxing Zhou
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Chengmin Zhang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Hu Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Xiaoming Cheng
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Xuemei Liu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China.
| | - Taolang Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China.
| |
Collapse
|
14
|
Low thyroid hormone receptor alpha-2 (THRα-2) tumor expression is associated with unfavorable tumor characteristics and high breast cancer mortality. Breast Cancer Res 2021; 23:117. [PMID: 34930399 PMCID: PMC8691018 DOI: 10.1186/s13058-021-01496-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 12/13/2021] [Indexed: 11/10/2022] Open
Abstract
Background The active thyroid hormone triiodothyronine (T3) has been found to have an estrogen-like effect on breast cancer cells. Thyroid hormone receptor alpha-2 (THRα-2) acts as an antagonist for triiodothyronine (T3) signaling, and a low expression has been associated with unfavorable tumor characteristics and a higher mortality in breast cancer. However, the evidence are not conclusive. The present study evaluates tumor-specific THRα-2 expression in invasive breast cancers and its association with tumor characteristics and long-term mortality in a large population. Method The Malmö Diet and Cancer Study (MDCS), a population-based cohort in Sweden that included 17,035 women from 1991 to 1996, was used. Women diagnosed with breast cancer during 1991–2010 were eligible for inclusion. A tissue micro array was constructed from stored tumor material and stained for THRα-2 using immunohistochemistry. Tumors from 654 patients were scored regarding the intensity and the fraction of cells stained, then dichotomized into low or high expression. Date and cause of death were collected up until 2018-12-31. Tumor- and patient characteristics were available from the MDCS. Missing data was imputed using chained equations. Logistic regression was used to calculate odds ratios (ORs) with 95% confidence intervals (CIs) for low vs high expression of THRα-2 related to specific tumor factors. Mortality was evaluated with Kaplan–Meier curves and Cox regression, rendering hazard ratios (HRs). Analyses were also stratified for estrogen receptor (ER) status. Results We found strong evidence of an association between low THRα-2 and unfavorable tumor characteristics, including estrogen receptor negativity: OR 4.04 (95% CI 2.28–7.15) and tumor size > 20–50 mm: OR 2.20 (95% CI 1.39–3.49). We found evidence of increased breast cancer-specific mortality for women with low THRα-2, HR 1.38 (95% CI 0.96–1.99), which remained after adjusting for age at diagnosis, HR 1.48 (95% CI 1.03–2.14), but not after adjusting for relevant prognostic factors, HR 0.98 (95% CI 0.66–1.45). THRα-2 expression in ER-negative tumors had an inverse correlation with overall mortality, HR 0.27 (95% CI 0.11–0.65). Conclusion Low tumor-specific THRα-2 expression was in this study associated with prognostically unfavorable tumor characteristics and a higher mortality in breast cancer, but not independent from other prognostic factors. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-021-01496-7.
Collapse
|
15
|
Wargasetia TL, Ratnawati H, Widodo N, Widyananda MH. Bioinformatics Study of Sea Cucumber Peptides as Antibreast Cancer Through Inhibiting the Activity of Overexpressed Protein (EGFR, PI3K, AKT1, and CDK4). Cancer Inform 2021; 20:11769351211031864. [PMID: 34345161 PMCID: PMC8283226 DOI: 10.1177/11769351211031864] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/22/2021] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is the most common type of cancer in women globally. The overexpressed proteins, including EGFR, PI3K, AKT1, and CDK4, have a role in the growth of breast cancer cells. The 3D peptide structure of sea cucumber Cucumaria frondosa was modeled and then docked with EGFR, PI3K, AKT1, and CDK4 proteins using AutoDock Vina software. The docking result, which has the best binding affinity value, is continued with molecular dynamics simulation. The docking results showed that all peptides bind to the active sites of the four proteins. WPPNYQW and YDWRF peptides bind to proteins with lower binding affinity values than positive controls. The four proteins were in a stable state when complexed with the WPPNYQW peptide, which was seen from the RMSD and RMSF value. PI3K-YDWRF and AKT1-YDWRF complexes are stable, characterized by high RMSD values and increased volatility in several amino acids. WPPNYQW peptide has high potential as an antibreast cancer agent because it binds to the active sites of the four proteins with low binding affinity values and stable interactions. Meanwhile, the YDWRF peptide interacts with the four proteins with low binding affinity values, but the interaction is only stable on PI3K and AKT1 proteins.
Collapse
Affiliation(s)
| | - Hana Ratnawati
- Faculty of Medicine, Maranatha Christian University, Bandung, Indonesia
| | - Nashi Widodo
- Biology Department, Faculty of Mathematics and Natural Sciences, The University of Brawijaya, Malang, Indonesia
| | | |
Collapse
|
16
|
Rustamadji P, Wiyarta E, Bethania KA, Kusmardi K. Potential of AKT2 expression as a predictor of lymph-node metastasis in invasive breast carcinoma of no special type. J Pathol Transl Med 2021; 55:271-278. [PMID: 34111909 PMCID: PMC8353139 DOI: 10.4132/jptm.2021.04.26] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/11/2021] [Accepted: 04/26/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Invasive breast carcinoma of no special type (IBC-NST) is the most common type of breast cancer and mainly causes regional lymph-node metastasis (LNM). We investigated the potential for AKT2 expression as a predictive biomarker for LNM in IBC-NST. METHODS Forty-eight paraffin blocks containing IBC-NST primary tumors were divided into two groups based on presence or absence of LNM. Age, tumor grade, tumor size, lymphovascular invasion (LVI), and AKT expression were assessed. AKT2 expression was assessed based on immunohistochemical staining, while other data were collected from archives. RESULTS Multiple logistic regression results showed that AKT2 expression and LVI were significantly associated with LNM (odds ratio [OR], 5.32; 95% confidence interval [CI], 1.42 to 19.93 and OR, 4.46; 95% CI, 1.17 to 16.97, respectively). AKT2 expression was able to discriminate against LNM (area under the receiver operating characteristic, 0.799 ± 0.063; 95% CI, 0.676 to 0.921) at an H-score cutoff of 104.62 (83.3% sensitivity, 62.5% specificity). CONCLUSIONS AKT2 expression has potential as a predictor of LNM in IBC-NST. The H-score cutoff for AKT2 expression can be used as a classification guide in future studies.
Collapse
Affiliation(s)
- Primariadewi Rustamadji
- Department of Anatomic Pathology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Elvan Wiyarta
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Kristina Anna Bethania
- Department of Anatomic Pathology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Kusmardi Kusmardi
- Department of Anatomic Pathology, Drug Development Research Cluster, Human Cancer Research Center, IMERI, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
17
|
Al-Bahlani SM, Lakhtakia R, Al-Jaaidi SS, Al-Sinawi SS, Abd-Elmoety SG, Al-Khabori M, Osman AHA, Al-Baimani K, Shalaby AA. Correlation of expression of Akt1 and E2F1 and their phosphorylated forms in breast cancer patients with clinicopathological parameters. J Mol Histol 2021; 52:621-633. [PMID: 33847878 DOI: 10.1007/s10735-021-09973-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/03/2021] [Indexed: 12/24/2022]
Abstract
Breast cancer is the leading cancer worldwide among women. Traditional clinicopathological prognostic and predictive markers need refining to improve clinical outcomes. This study explored the association between traditional clinicopathological factors and the expression of Akt1 and E2F1 transduction proteins and their phosphorylated forms in breast cancer, to determine their value as novel biomarkers and potential therapeutic targets. Tumor tissues from 94 female breast cancer patients were examined for immunophenotypic expression of total Akt1, pAkt1 (Serine 473), pAkt1 (Threonine 308), total E2F1, pE2F1 (Thr433) and pE2F1 (Ser337). The expression of pAkt1 (Ser473) was significantly associated with ER/PR positive status and total E2F1 with older age (> 50), lymph node involvement and HER2 positivity. There was a significant association between triple negative cancers and total and pAkt1 (Thr308). pAkt1 (Ser473) showed an inverse relationship with Luminal B cancers and pE2F1 (Thr433) showed an inverse association with triple negative cancers. Higher expression of pE2F1 (Ser337) was associated with better OS. Both pAkt1 (Ser473 and Thr308) proteins showed significant association with poorer patient outcomes. E2F1 (Ser337) showed a significant positive correlation with response to chemotherapy. The study suggests that a pAkt1-/pE2F1+ phenotype could indicate an opportunity to minimize chemotherapeutic options in older women; conversely a pAkt1+/pE2F1- phenotype could prompt a more aggressive regimen. Further exploration of this phenotype in younger women with breast cancer and triple-negative breast cancers is warranted.
Collapse
Affiliation(s)
- Shadia M Al-Bahlani
- Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, Alkhoud, PC 123, P.O. Box 35, Muscat, Oman.
| | - Ritu Lakhtakia
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health Care City, Dubai, UAE
| | - Samiya S Al-Jaaidi
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Shadia S Al-Sinawi
- Department of Pathology, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat, Oman
| | - Shaymaa G Abd-Elmoety
- Department of Pathology, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat, Oman
| | - Murtadha Al-Khabori
- Department of Hematology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Anjum H A Osman
- Oncology Unit, Department of Medicine, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat, Oman
| | - Khalid Al-Baimani
- Oncology Unit, Department of Medicine, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat, Oman
| | - Asem A Shalaby
- Department of Pathology, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat, Oman
- Pathology Department, College of Medicine, Mansoura University, El Mansûra, Egypt
| |
Collapse
|
18
|
Popova NV, Jücker M. The Role of mTOR Signaling as a Therapeutic Target in Cancer. Int J Mol Sci 2021; 22:ijms22041743. [PMID: 33572326 PMCID: PMC7916160 DOI: 10.3390/ijms22041743] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 01/30/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
The aim of this review was to summarize current available information about the role of phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling in cancer as a potential target for new therapy options. The mTOR and PI3K/AKT/mTORC1 (mTOR complex 1) signaling are critical for the regulation of many fundamental cell processes including protein synthesis, cell growth, metabolism, survival, catabolism, and autophagy, and deregulated mTOR signaling is implicated in cancer, metabolic dysregulation, and the aging process. In this review, we summarize the information about the structure and function of the mTOR pathway and discuss the mechanisms of its deregulation in human cancers including genetic alterations of PI3K/AKT/mTOR pathway components. We also present recent data regarding the PI3K/AKT/mTOR inhibitors in clinical studies and the treatment of cancer, as well the attendant problems of resistance and adverse effects.
Collapse
Affiliation(s)
- Nadezhda V. Popova
- Laboratory of Receptor Cell Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str., 16/10, 117997 Moscow, Russia;
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
- Correspondence: ; Tel.: +49-(0)-40-7410-56339
| |
Collapse
|
19
|
Mechanism of action biomarkers predicting response to AKT inhibition in the I-SPY 2 breast cancer trial. NPJ Breast Cancer 2020; 6:48. [PMID: 33083527 PMCID: PMC7532145 DOI: 10.1038/s41523-020-00189-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
The AKT inhibitor MK2206 (M) was evaluated in I-SPY 2 and graduated in
the HER2+, HR−, and HR− HER2+ signatures. We hypothesized that AKT signaling axis
proteins/genes may specifically predict response to M and tested 26 phospho-proteins
and 10 genes involved in AKT-mTOR-HER signaling; in addition, we tested 9 genes from
a previous study in the metastatic setting. One hundred and fifty patients had gene
expression data from pretreatment biopsies available for analysis (M: 94, control:
56) and 138 had protein data (M: 87, control: 51). Logistic modeling was used to
assess biomarker performance in pre-specified analysis. In general, phospho-protein
biomarkers of activity in the AKT-mTOR-HER pathway appeared more predictive of
response to M than gene expression or total protein biomarkers in the same pathway;
however, the nature of the predictive biomarkers differed in the HER2+ and TN
groups. In the HER2+ subset, patients achieving a pCR in M had higher levels of
multiple AKT kinase substrate phospho-proteins (e.g., pmTOR, pTSC2). In contrast, in
the TN subset responding patients had lower levels of AKT pathway phospho-proteins,
such as pAKT, pmTOR, and pTSC2. Pathway mutations did not appear to account for
these associations. Additional exploratory whole-transcriptome analysis revealed
immune signaling as strongly associated with response to M in the HER2+ subset.
While our sample size is small, these results suggest that the measurement of
particular AKT kinase substrate phospho-proteins could be predictive of MK2206
efficacy in both HER2+ and TN tumors and that immune signaling may play a role in
response in HER2+ patients.
Collapse
|
20
|
Huang G, Song C, Wang N, Qin T, Sui S, Obr A, Zeng L, Wood TL, Leroith D, Li M, Wu Y. RNA-binding protein CUGBP1 controls the differential INSR splicing in molecular subtypes of breast cancer cells and affects cell aggressiveness. Carcinogenesis 2020; 41:1294-1305. [PMID: 31958132 PMCID: PMC7513956 DOI: 10.1093/carcin/bgz141] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/18/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
The insulin receptor gene (INSR) undergoes alternative splicing to give rise to two functionally related, but also distinct, isoforms IR-A and IR-B, which dictate proliferative and metabolic regulations, respectively. Previous studies identified the RNA-binding protein CUGBP1 as a key regulator of INSR splicing. In this study, we show that the differential splicing of INSR occurs more frequently in breast cancer than in non-tumor breast tissues. In breast cancer cell lines, the IR-A:IR-B ratio varies in different molecular subtypes, knockdown or overexpression of CUGBP1 gene in breast cancer cells altered IR-A:IR-B ratio through modulation of IR-A expression, thereby reversed or enhanced the insulin-induced oncogenic behavior of breast cancer cells, respectively. Our data revealed the predominant mitogenic role of IR-A isoform in breast cancer and depicted a novel interplay between INSR and CUGBP1, implicating CUGBP1 and IR-A isoform as the potential therapeutic targets and biomarkers for breast cancer.
Collapse
Affiliation(s)
- Gena Huang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Chen Song
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Tao Qin
- Department of Pathology, Dalian Medical University, Dalian, Liaoning, China
| | - Silei Sui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Alison Obr
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Li Zeng
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Derek Leroith
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
| | - Man Li
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yingjie Wu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
- College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
21
|
Abdallah ME, El-Readi MZ, Althubiti MA, Almaimani RA, Ismail AM, Idris S, Refaat B, Almalki WH, Babakr AT, Mukhtar MH, Abdalla AN, Idris OF. Tamoxifen and the PI3K Inhibitor: LY294002 Synergistically Induce Apoptosis and Cell Cycle Arrest in Breast Cancer MCF-7 Cells. Molecules 2020; 25:E3355. [PMID: 32722075 PMCID: PMC7436112 DOI: 10.3390/molecules25153355] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is considered as one of the most aggressive types of cancer. Acquired therapeutic resistance is the major cause of chemotherapy failure in breast cancer patients. To overcome this resistance and to improve the efficacy of treatment, drug combination is employed as a promising approach for this purpose. The synergistic cytotoxic, apoptosis inducing, and cell cycle effects of the combination of LY294002 (LY), a phosphatidylinositide-3-kinase (PI3K) inhibitor, with the traditional cytotoxic anti-estrogen drug tamoxifen (TAM) in breast cancer cells (MCF-7) were investigated. LY and TAM exhibited potent cytotoxic effect on MCF-7 cells with IC50 values 0.87 µM and 1.02 µM. The combination of non-toxic concentration of LY and TAM showed highly significant synergistic interaction as observed from isobologram (IC50: 0.17 µM, combination index: 0.18, colony formation: 9.01%) compared to untreated control. The percentage of early/late apoptosis significantly increased after treatment of MCF-7 cells with LY and TAM combination: 40.3%/28.3% (p < 0.001), compared to LY single treatment (19.8%/11.4%) and TAM single treatment (32.4%/5.9%). In addition, LY and TAM combination induced the apoptotic genes Caspase-3, Caspase-7, and p53, as well as p21 as cell cycle promotor, and significantly downregulated the anti-apoptotic genes Bcl-2 and survivin. The cell cycle assay revealed that the combination induced apoptosis by increasing the pre-G1: 28.3% compared to 1.6% of control. pAKT and Cyclin D1 protein expressions were significantly more downregulated by the combination treatment compared to the single drug treatment. The results suggested that the synergistic cytotoxic effect of LY and TAM is achieved by the induction of apoptosis and cell cycle arrest through cyclin D1, pAKT, caspases, and Bcl-2 signaling pathways.
Collapse
Affiliation(s)
- Mohamed E. Abdallah
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Mahmoud Zaki El-Readi
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Mohammad Ahmad Althubiti
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Riyad Adnan Almaimani
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Amar Mohamed Ismail
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, Al-Neelain University, Khartoum 11121, Sudan; (A.M.I.); (O.F.I.)
| | - Shakir Idris
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 7607, Saudi Arabia; (S.I.); (B.R.)
| | - Bassem Refaat
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 7607, Saudi Arabia; (S.I.); (B.R.)
| | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Abdullatif Taha Babakr
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Mohammed H. Mukhtar
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Ashraf N. Abdalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
- Department of Pharmacology and Toxicology, Medicinal and Aromatic Plants Research Institute, National Center for Research, Khartoum 2404, Sudan
| | - Omer Fadul Idris
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, Al-Neelain University, Khartoum 11121, Sudan; (A.M.I.); (O.F.I.)
| |
Collapse
|
22
|
Roldán DB, Grimmler M, Hartmann C, Hubich-Rau S, Beißert T, Paret C, Cagna G, Rohde C, Wöll S, Koslowski M, Türeci Ö, Sahin U. PLAC1 is essential for FGF7/FGFRIIIb-induced Akt-mediated cancer cell proliferation. Oncotarget 2020; 11:1862-1875. [PMID: 32499871 PMCID: PMC7244013 DOI: 10.18632/oncotarget.27582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/14/2020] [Indexed: 12/20/2022] Open
Abstract
PLAC1 (placenta enriched 1) is a mammalian trophoblast-specific protein. Aberrant expression of PLAC1 is observed in various human cancers, where it is involved in the motility, migration, and invasion of tumor cells, which are associated with the phosphoinositide 3-kinase (PI3K)/AKT pathway. We previously demonstrated that AKT activation mediates the downstream effects of PLAC1; however, the molecular mechanisms of PLAC1-induced AKT-mediated tumor-related processes are unclear. We studied human choriocarcinoma and breast cancer cell lines to explore the localization and receptor-ligand interactions, as well as the downstream effects of PLAC1. We show secretion and adherence of PLAC1 to the extracellular matrix, where it forms a trimeric complex with fibroblast growth factor 7 (FGF7) and its receptor, FGF receptor 2 IIIb (FGFR2IIIb). We further show that PLAC1 signaling via FGFR2IIIb activates AKT phosphorylation in cancer cell lines. As the FGF pathway is of major interest in anticancer therapeutic strategies, these data further promote PLAC1 as a promising anticancer drug target.
Collapse
Affiliation(s)
- Diana Barea Roldán
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- These authors contributed equally to this work
| | - Matthias Grimmler
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: DiaSys Diagnostic Systems GmbH, Holzheim, Germany
- These authors contributed equally to this work
| | - Christoph Hartmann
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: Merck KGaA, Darmstadt, Germany
- These authors contributed equally to this work
| | - Stefanie Hubich-Rau
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- These authors contributed equally to this work
| | - Tim Beißert
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Paret
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Giuseppe Cagna
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Current address: Lonza Pharma & Biotech, Cologne, Germany
| | - Christoph Rohde
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Current address: Merck KGaA, Darmstadt, Germany
| | - Stefan Wöll
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Michael Koslowski
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Formerly of University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: GammaDelta Therapeutics, London, United Kingdom
| | - Özlem Türeci
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
- Ci3 Cluster for Individualized Immune Intervention, Mainz, Germany
| | - Ugur Sahin
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
23
|
Kruger DT, Opdam M, Sanders J, van der Noort V, Boven E, Linn SC. Hierarchical clustering of PI3K and MAPK pathway proteins in breast cancer intrinsic subtypes. APMIS 2020; 128:298-307. [PMID: 31976581 PMCID: PMC7317370 DOI: 10.1111/apm.13026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 01/10/2020] [Indexed: 01/04/2023]
Abstract
The phosphatidylinositol‐3‐kinase (PI3K) and mitogen‐activated protein kinase (MAPK) pathways are frequently activated in breast cancer. We recently demonstrated the importance of analyzing multiple proteins as read‐out for pathway activation in ER+/HER2− breast cancer, since single proteins are known to provide insufficient information. Here, we determined pathway activation in other primary breast cancer intrinsic subtypes derived from postmenopausal patients. Tumor blocks were recollected, and immunohistochemistry was performed using antibodies against PTEN, p‐AKT(Thr308), p‐AKT(Ser473), p‐p70S6K, p‐4EBP1, p‐S6RP(Ser235/236) and p‐ERK1/2, followed by unsupervised hierarchical clustering. In 32 ER+/HER2+, 37 ER−/HER2+ and 74 triple‐negative breast cancer patients, subgroups were identified with preferentially activated (A) and preferentially not activated (N) proteins. These subgroups likely reflect tumors with differences in biological behavior as well as treatment outcome.
Collapse
Affiliation(s)
- Dinja T Kruger
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, The Netherlands.,Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Vincent van der Noort
- Division of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Epie Boven
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sabine C Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Pathology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
24
|
Iida M, Harari PM, Wheeler DL, Toulany M. Targeting AKT/PKB to improve treatment outcomes for solid tumors. Mutat Res 2020; 819-820:111690. [PMID: 32120136 DOI: 10.1016/j.mrfmmm.2020.111690] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/31/2020] [Accepted: 02/11/2020] [Indexed: 12/16/2022]
Abstract
The serine/threonine kinase AKT, also known as protein kinase B (PKB), is the major substrate to phosphoinositide 3-kinase (PI3K) and consists of three paralogs: AKT1 (PKBα), AKT2 (PKBβ) and AKT3 (PKBγ). The PI3K/AKT pathway is normally activated by binding of ligands to membrane-bound receptor tyrosine kinases (RTKs) as well as downstream to G-protein coupled receptors and integrin-linked kinase. Through multiple downstream substrates, activated AKT controls a wide variety of cellular functions including cell proliferation, survival, metabolism, and angiogenesis in both normal and malignant cells. In human cancers, the PI3K/AKT pathway is most frequently hyperactivated due to mutations and/or overexpression of upstream components. Aberrant expression of RTKs, gain of function mutations in PIK3CA, RAS, PDPK1, and AKT itself, as well as loss of function mutation in AKT phosphatases are genetic lesions that confer hyperactivation of AKT. Activated AKT stimulates DNA repair, e.g. double strand break repair after radiotherapy. Likewise, AKT attenuates chemotherapy-induced apoptosis. These observations suggest that a crucial link exists between AKT and DNA damage. Thus, AKT could be a major predictive marker of conventional cancer therapy, molecularly targeted therapy, and immunotherapy for solid tumors. In this review, we summarize the current understanding by which activated AKT mediates resistance to cancer treatment modalities, i.e. radiotherapy, chemotherapy, and RTK targeted therapy. Next, the effect of AKT on response of tumor cells to RTK targeted strategies will be discussed. Finally, we will provide a brief summary on the clinical trials of AKT inhibitors in combination with radiochemotherapy, RTK targeted therapy, and immunotherapy.
Collapse
Affiliation(s)
- M Iida
- Department of Human Oncology, University of Wisconsin in Madison, Madison, WI, USA.
| | - P M Harari
- Department of Human Oncology, University of Wisconsin in Madison, Madison, WI, USA
| | - D L Wheeler
- Department of Human Oncology, University of Wisconsin in Madison, Madison, WI, USA
| | - M Toulany
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany; German Cancer Consortium (DKTK), Partner Site Tuebingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
25
|
Vasheghani Farahani M, Ataee Dizaji P, Rashidi H, Mokarian F, Biglarian A. Application of Multi-State Model in Analyzing of Breast Cancer Data. J Res Health Sci 2020; 19:e00465. [PMID: 32291364 PMCID: PMC7183561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 12/27/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The multistate model is used generally to fit the longitudinal data. This model can determine the natural trend of disease progress in different states of treatment, recuperate, metastasis and finally death. We aimed to use multistate models in order to analyzing breast cancer (BC) data. STUDY DESIGN A historical cohort study. METHODS In this historical cohort study, 573 women with BC were studied. These patients were referred to Isfahan Sayed-o-Shohada Hospital during 1999-2006 and followed up to Apr 2017. The corresponding provided data were gathered by Isfahan Cancer Prevention Center. Then data analyzed by multistate models in R 3.4.1 software. RESULTS The mean and standard deviation of women age were 47.19±10.77 years. The transition probability from state of first treatment to recuperate state was 71%, to metastasis state 2% and to death was 16%. The sojourn time in different states of disease was 2.39 yr for first treatment, 6.93 yr for recuperate and 0.16 yr for death. CONCLUSION This model is able to predict the transition probabilities in different state of disease, so its results are useful for clinical researches. In addition, with transition probabilities and also survival mean in each state in hand, the physicians will be able to suggest suitable treatment plans for patients.
Collapse
Affiliation(s)
| | - Parisa Ataee Dizaji
- 1Department of Biostatistics, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Hamid Rashidi
- 1Department of Biostatistics, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Fariborz Mokarian
- 2Faculty of Medicine, Cancer Prevention Research Center, Isfahan University of Medical Sciences, Iran
| | - Akbar Biglarian
- 3Department of Biostatistics, Social Determinants of Health Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran,Correspondence : Akbar Biglarian (PhD) Tel: +98(21)022180146 E-mail:
| |
Collapse
|
26
|
Yee LD, Mortimer JE, Natarajan R, Dietze EC, Seewaldt VL. Metabolic Health, Insulin, and Breast Cancer: Why Oncologists Should Care About Insulin. Front Endocrinol (Lausanne) 2020; 11:58. [PMID: 32153503 PMCID: PMC7045050 DOI: 10.3389/fendo.2020.00058] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
Studies investigating the potential link between adult pre-menopausal obesity [as measured by body mass index (BMI)] and triple-negative breast cancer have been inconsistent. Recent studies show that BMI is not an exact measure of metabolic health; individuals can be obese (BMI > 30 kg/m2) and metabolically healthy or lean (BMI < 25 kg/m2) and metabolically unhealthy. Consequently, there is a need to better understand the molecular signaling pathways that might be activated in individuals that are metabolically unhealthy and how these signaling pathways may drive biologically aggressive breast cancer. One key driver of both type-2 diabetes and cancer is insulin. Insulin is a potent hormone that activates many pathways that drive aggressive breast cancer biology. Here, we review (1) the controversial relationship between obesity and breast cancer, (2) the impact of insulin on organs, subcellular components, and cancer processes, (3) the potential link between insulin-signaling and cancer, and (4) consider time points during breast cancer prevention and treatment where insulin-signaling could be better controlled, with the ultimate goal of improving overall health, optimizing breast cancer prevention, and improving breast cancer survival.
Collapse
|
27
|
The exosome-mediated autocrine and paracrine actions of plasma gelsolin in ovarian cancer chemoresistance. Oncogene 2019; 39:1600-1616. [PMID: 31700155 PMCID: PMC7018662 DOI: 10.1038/s41388-019-1087-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 02/06/2023]
Abstract
Ovarian cancer (OVCA) is the most lethal gynecological cancer, due predominantly to late presentation, high recurrence rate and common chemoresistance development. The expression of the actin-associated protein cytosolic gelsolin (GSN) regulates the gynecological cancer cell fate resulting in dysregulation in chemosensitivity. In this study, we report that elevated expression of plasma gelsolin (pGSN), a secreted isoform of GSN and expressed from the same GSN gene, correlates with poorer overall survival and relapse-free survival in patients with OVCA. In addition, it is highly expressed and secreted in chemoresistant OVCA cells than its chemosensitive counterparts. pGSN, secreted and transported via exosomes (Ex-pGSN), upregulates HIF1α–mediated pGSN expression in chemoresistant OVCA cells in an autocrine manner as well as confers cisplatin resistance in otherwise chemosensitive OVCA cells. These findings support our hypothesis that exosomal pGSN promotes OVCA cell survival through both autocrine and paracrine mechanisms that transform chemosensitive cells to resistant counterparts. Specifically, pGSN transported via exosomes is a determinant of chemoresistance in OVCA.
Collapse
|
28
|
Wu Z, Wu J, Zhao Q, Fu S, Jin J. Emerging roles of aerobic glycolysis in breast cancer. Clin Transl Oncol 2019; 22:631-646. [PMID: 31359335 DOI: 10.1007/s12094-019-02187-8] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/05/2019] [Indexed: 12/25/2022]
Abstract
Altered aerobic glycolysis is a well-recognized characteristic of cancer cell energy metabolism, known as the Warburg effect. Even in the presence of abundant oxygen, a majority of tumor cells produce substantial amounts of energy through a high glycolytic metabolism, and breast cancer (BC) is no exception. Breast cancer continues to be the second leading cause of cancer-associated mortality in women worldwide. However, the precise role of aerobic glycolysis in the development of BC remains elusive. Therefore, the present review attempts to address the implication of key enzymes of the aerobic glycolytic pathway including hexokinase (HK), phosphofructokinase (PFK) and pyruvate kinase (PK), glucose transporters (GLUTs), together with related signaling pathways including protein kinase B(PI3K/AKT), mammalian target of rapamycin (mTOR) and adenosine monophosphate-activated protein kinase (AMPK) and transcription factors (c-myc, p53 and HIF-1) in the research of BC. Thus, the review of aerobic glycolysis in BC may evoke novel ideas for the BC treatment.
Collapse
Affiliation(s)
- Z Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - J Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Q Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, People's Republic of China
| | - S Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| | - J Jin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| |
Collapse
|
29
|
Quintanal-Villalonga A, Ferrer I, Molina-Pinelo S, Paz-Ares L. A patent review of FGFR4 selective inhibition in cancer (2007-2018). Expert Opin Ther Pat 2019; 29:429-438. [DOI: 10.1080/13543776.2019.1624720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
| | - Irene Ferrer
- CNIO-H12O Lung Clinical Cancer Research Unit, Fundación de Investigación Biomédica i+12 & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Sonia Molina-Pinelo
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
| | - Luis Paz-Ares
- CNIO-H12O Lung Clinical Cancer Research Unit, Fundación de Investigación Biomédica i+12 & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Doce de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Medical School, Universidad Complutense, Madrid, Spain
| |
Collapse
|
30
|
Chantarawong W, Kuncharoen N, Tanasupawat S, Chanvorachote P. Lumichrome Inhibits Human Lung Cancer Cell Growth and Induces Apoptosis via a p53-Dependent Mechanism. Nutr Cancer 2019; 71:1390-1402. [PMID: 31074646 DOI: 10.1080/01635581.2019.1610183] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Lumichrome, a major derivative of riboflavin, may exhibit pharmacological activity against cancer cells. Riboflavin is a vitamin found in food, however, certain evidence has suggested its possible potentiating effects on cancer progression. Here, we have shown for the first time that unlike riboflavin, lumichrome can suppress lung cancer cell growth and reduce survival in both normal and anchorage-independent conditions. In addition, lumichrome induced apoptosis in lung cancer cells via a p53-dependent mitochondrial mechanism with substantial selectivity, shown by its lesser toxicity to the normal primary dermal papilla cells. The potency of lumichrome in killing lung cancer cells was found to be comparable to that of cisplatin, a standard chemotherapeutic drug for lung cancer treatment. With regard to the mechanism, lumichrome significantly upregulated p53 and decreased its downstream target BCL-2. Such a shift of BCL-2 family protein balance further activated caspase-9 and -3 and finally executed apoptosis. Furthermore, lumichrome potentially suppressed cancer stem cells (CSCs) in lung cancer by dramatically suppressing CSC markers together with the CSC-maintaining cell signaling namely protein kinase B (AKT) and β-catenin. To conclude, the present study has unraveled a novel role and mechanism of lumichrome against lung cancer that may benefit the development of the compound for management of the disease.
Collapse
Affiliation(s)
- Wipa Chantarawong
- Cell-based Drug and Health Products Development Research Unit, Chulalongkorn University , Bangkok , Thailand
| | - Nattakorn Kuncharoen
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University , Bangkok , Thailand
| | - Somboon Tanasupawat
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University , Bangkok , Thailand
| | - Pithi Chanvorachote
- Cell-based Drug and Health Products Development Research Unit, Chulalongkorn University , Bangkok , Thailand.,Departments of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University , Bangkok , Thailand
| |
Collapse
|
31
|
Li X, Fu Q, Li H, Zhu L, Chen W, Ruan T, Xu W, Yu X. MicroRNA‐520c‐3p functions as a novel tumor suppressor in lung adenocarcinoma. FEBS J 2019; 286:2737-2752. [PMID: 30942957 DOI: 10.1111/febs.14835] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/28/2019] [Accepted: 04/01/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital China
- National Clinical Research Center for Cancer Tianjin China
- Key Laboratory of Cancer Prevention and Therapy Tianjin China
- Tianjin's Clinical Research Center for Cancer China
| | - Qiang Fu
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital China
- National Clinical Research Center for Cancer Tianjin China
- Key Laboratory of Cancer Prevention and Therapy Tianjin China
- Tianjin's Clinical Research Center for Cancer China
| | - Hui Li
- National Clinical Research Center for Cancer Tianjin China
- Tianjin's Clinical Research Center for Cancer China
- Key Laboratory of Cancer Immunology and Biotherapy Tianjin China
- Department of Gastrointestinal Cancer Biology Tianjin Medical University Cancer Institute and Hospital China
| | - Lei Zhu
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital China
- National Clinical Research Center for Cancer Tianjin China
- Key Laboratory of Cancer Prevention and Therapy Tianjin China
- Tianjin's Clinical Research Center for Cancer China
| | - Wei Chen
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital China
- National Clinical Research Center for Cancer Tianjin China
- Key Laboratory of Cancer Prevention and Therapy Tianjin China
- Tianjin's Clinical Research Center for Cancer China
| | - Tonglei Ruan
- Department of Gynecology and Obstetrics Tianjin Medical University General Hospital Tianjin China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital China
- National Clinical Research Center for Cancer Tianjin China
- Key Laboratory of Cancer Prevention and Therapy Tianjin China
- Tianjin's Clinical Research Center for Cancer China
| | - Xiaozhou Yu
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital China
- National Clinical Research Center for Cancer Tianjin China
- Key Laboratory of Cancer Prevention and Therapy Tianjin China
- Tianjin's Clinical Research Center for Cancer China
| |
Collapse
|
32
|
Ahmed S, Khan H, Fratantonio D, Hasan MM, Sharifi S, Fathi N, Ullah H, Rastrelli L. Apoptosis induced by luteolin in breast cancer: Mechanistic and therapeutic perspectives. PHYTOMEDICINE 2019; 59:152883. [PMID: 30986716 DOI: 10.1016/j.phymed.2019.152883] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/06/2019] [Accepted: 03/09/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Breast cancer is worldwide commonly found malignancy in women and effective treatment is regarded as a huge clinical challenge even in the presence of several options. Extensive literature is available that demonstrating polyphenols, the richly introduce phytopharmaceuticals as anticancer agents. Among these polyphenols, resveratrol, silibinin, quercetin, genistein, curcumin reported to have an awesome potential against breast cancer. However, till now no comprehensive survey found about the anticarcinogenic properties of luteolin against breast cancer. SCOPE AND APPROACH This review targeted the available literature on luteolin in the treatment of breast cancer, effects in combination with other anticancer drugs with possible mechanisms. KEY FINDINGS AND CONCLUSION An outstanding therapeutic potential of luteolin in the treatment of breast cancer has been recorded not just as a chemopreventive and chemotherapeutic agent yet complemented by its synergistic effects with other anticancer therapies such as cyclophosphamide, doxorubicin, and NSAID such as celecoxib, and possible underlying mechanisms. Ideally, this review will open new dimensions for luteolin as an effective and safe therapeutic agent in diminishing breast cancer.
Collapse
Affiliation(s)
- Salman Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali khan University Mardan 23200, Pakistan.
| | - Deborah Fratantonio
- "Bambino Gesù" Children's Hospital-IRCCS, Research Laboratories, V.le di San Paolo 15, 00146, Rome, Italy.
| | - Muhammad Mohtasheemul Hasan
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazanin Fathi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hammad Ullah
- Department of Pharmacy, Abdul Wali khan University Mardan 23200, Pakistan
| | - Luca Rastrelli
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II, 84084, Fisciano, Italy
| |
Collapse
|
33
|
Quintanal-Villalonga A, Molina-Pinelo S, Yagüe P, Marrugal Á, Ojeda-Márquez L, Suarez R, Ponce-Aix S, Enguita AB, Carnero A, Ferrer I, Paz-Ares L. FGFR4 increases EGFR oncogenic signaling in lung adenocarcinoma, and their combined inhibition is highly effective. Lung Cancer 2019; 131:112-121. [PMID: 31027687 DOI: 10.1016/j.lungcan.2019.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/04/2019] [Accepted: 02/07/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Lung adenocarcinoma accounts for approximately half of lung cancer cases. Twenty to 50% of tumors of this type harbor mutations affecting epidermal growth factor receptor (EGFR) expression or activity, which can be therapeutically targeted. EGFR inhibitors in this context exhibit high efficacy and are currently used in the clinical setting. However, not all adenocarcinomas harboring EGFR mutations respond to therapy, so predictive biomarkers of therapeutic outcomes, as well as novel therapies sensitizing these tumors to EGFR inhibition, are needed. MATERIALS AND METHODS We performed in vitro gene overexpression/silencing and tumorigenic surrogate assays, as well as in vitro and in vivo combination treatments with Fibroblast Growth Factor Receptor (FGFR)/EGFR inhibitors. At the clinical level, we determined FGFR4 expression levels in tumors from patients treated with EGFR inhibitors and correlated these with treatment response. RESULTS We describe a cooperative interaction between EGFR and FGFR4, which results in their reciprocal activation with pro-oncogenic consequences in vitro and in vivo. This cooperation is independent of EGFR activating mutations and increases resistance to different EGFR inhibitors. At the therapeutic level, we provide evidence of the synergistic effects of the combination of EGFR and FGFR inhibitors in high FGFR4-expressing, EGFR-activated tumors in vitro and in vivo. Correlated with these results, we found that patients treated with EGFR inhibitors relapse earlier when their tumors exhibit high FGFR4 expression. CONCLUSIONS We propose a novel predictive biomarker for EGFR-targeted therapy, and a highly efficacious combinatory therapeutic strategy to treat EGFR-dependent; this may may extend the use of appropriate inhibitors beyond EGFR-mutated adenocarcinoma patients.
Collapse
Affiliation(s)
- Alvaro Quintanal-Villalonga
- H120-CNIO Lung Cancer Clinical Cancer Research Unit, Fundación de Investigación Biomédica i+12 & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York, United States
| | - Sonia Molina-Pinelo
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain; CIBERONC, Madrid, Spain
| | - Patricia Yagüe
- H120-CNIO Lung Cancer Clinical Cancer Research Unit, Fundación de Investigación Biomédica i+12 & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; CIBERONC, Madrid, Spain
| | - Ángela Marrugal
- H120-CNIO Lung Cancer Clinical Cancer Research Unit, Fundación de Investigación Biomédica i+12 & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Laura Ojeda-Márquez
- H120-CNIO Lung Cancer Clinical Cancer Research Unit, Fundación de Investigación Biomédica i+12 & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; CIBERONC, Madrid, Spain
| | - Rocío Suarez
- H120-CNIO Lung Cancer Clinical Cancer Research Unit, Fundación de Investigación Biomédica i+12 & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; CIBERONC, Madrid, Spain
| | - Santiago Ponce-Aix
- Medical Oncology Department, Hospital Universitario Doce de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; CIBERONC, Madrid, Spain
| | - Ana Belén Enguita
- Pathological Anatomy Department, Hospital Universitario Doce de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain; CIBERONC, Madrid, Spain
| | - Irene Ferrer
- H120-CNIO Lung Cancer Clinical Cancer Research Unit, Fundación de Investigación Biomédica i+12 & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; CIBERONC, Madrid, Spain.
| | - Luis Paz-Ares
- H120-CNIO Lung Cancer Clinical Cancer Research Unit, Fundación de Investigación Biomédica i+12 & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; Medical Oncology Department, Hospital Universitario Doce de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; Medical School, Universidad Complutense, Madrid, Spain; CIBERONC, Madrid, Spain.
| |
Collapse
|
34
|
Akt1 inhibition promotes breast cancer metastasis through EGFR-mediated β-catenin nuclear accumulation. Cell Commun Signal 2018; 16:82. [PMID: 30445978 PMCID: PMC6240210 DOI: 10.1186/s12964-018-0295-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/06/2018] [Indexed: 12/11/2022] Open
Abstract
Background Knockdown of Akt1 promotes Epithelial-to-Mesenchymal Transition in breast cancer cells. However, the mechanisms are not completely understood. Methods Western blotting, immunofluorescence, luciferase assay, real time PCR, ELISA and Matrigel invasion assay were used to investigate how Akt1 inhibition promotes breast cancer cell invasion in vitro. Mouse model of lung metastasis was used to measure in vivo efficacy of Akt inhibitor MK2206 and its combination with Gefitinib. Results Knockdown of Akt1 stimulated β-catenin nuclear accumulation, resulting in breast cancer cell invasion. β-catenin nuclear accumulation induced by Akt1 inhibition depended on the prolonged activation of EGFR signaling pathway in breast cancer cells. Mechanistic experiments documented that knockdown of Akt1 inactivates PIKfyve via dephosphorylating of PIKfyve at Ser318 site, resulting in a decreased degradation of EGFR signaling pathway. Inhibition of Akt1 using MK2206 could induce an increase in the expression of EGFR and β-catenin in breast cancer cells. In addition, MK2206 at a low dosage enhance breast cancer metastasis in a mouse model of lung metastasis, while an inhibitor of EGFR tyrosine kinase Gefitinib could potentially suppress breast cancer metastasis induced by Akt1 inhibition. Conclusion EGFR-mediated β-catenin nuclear accumulation is critical for Akt1 inhibition-induced breast cancer metastasis.
Collapse
|
35
|
Kruger DT, Beelen KJ, Opdam M, Sanders J, van der Noort V, Boven E, Linn SC. Hierarchical clustering of activated proteins in the PI3K and MAPK pathways in ER-positive, HER2-negative breast cancer with potential therapeutic consequences. Br J Cancer 2018; 119:832-839. [PMID: 30287915 PMCID: PMC6189147 DOI: 10.1038/s41416-018-0221-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/11/2022] Open
Abstract
Background The phosphatidylinositol-3-kinase (PI3K) and/or mitogen-activated protein kinase (MAPK) pathways are frequently activated in breast cancer which can result in antioestrogen resistance. Single protein markers failed to be introduced into clinical practice. We, therefore, aimed to find a better read-out of activation of the PI3K and MAPK pathways in ER+/HER2− breast cancer. Assessment of seven PI3K/MAPK proteins might better reflect pathway activation and distinguish patients without adjuvant tamoxifen benefit. Methods Tumour blocks were recollected from 293 primary postmenopausal ER+/HER2− breast cancer patients randomised between tamoxifen and no adjuvant therapy. PTEN, p-AKT(Thr308), p-AKT(Ser473), p-p70S6K, p-4EBP1, p-ERK1/2 and p-S6RP expression was assessed by immunohistochemistry followed by unsupervised hierarchical clustering. The primary endpoint was recurrence-free interval. Multivariate Cox models were used to assess tamoxifen benefit. A classification tool was developed based on protein expression profile. Results Subgroups were identified with preferentially activated (A) and preferentially not activated (N) proteins. Patients in group N derived significant benefit from tamoxifen (multivariate hazard ratio (HR) = 0.23, p = 0.000101), while patients from group A did not (multivariate HR = 1.37, p = 0.64), p for interaction 0.020. Our generated classification tool confirmed these results (p for interaction 0.024). Conclusions Hierarchical clustering of seven PI3K/MAPK proteins reflects pathway activation and can guide treatment decisions in primary ER+/HER2− postmenopausal breast cancer patients.
Collapse
Affiliation(s)
- Dinja T Kruger
- Department of Medical Oncology, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Karin J Beelen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.,Department of Medical Oncology, Reinier de Graaf Groep, Reinier de Graafweg 5, 2625 AD, Delft, The Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Vincent van der Noort
- Division of Biometrics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Epie Boven
- Department of Medical Oncology, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Sabine C Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands. .,Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands. .,Department of Pathology, University Medical Centre Utrecht, and Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Sarwar M, Syed Khaja AS, Aleskandarany M, Karlsson R, Althobiti M, Ødum N, Mongan NP, Dizeyi N, Johnson H, Green AR, Ellis IO, Rakha EA, Persson JL. The role of PIP5K1α/pAKT and targeted inhibition of growth of subtypes of breast cancer using PIP5K1α inhibitor. Oncogene 2018; 38:375-389. [PMID: 30104711 PMCID: PMC6336681 DOI: 10.1038/s41388-018-0438-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/22/2018] [Accepted: 07/14/2018] [Indexed: 02/08/2023]
Abstract
Despite recent improvement in adjuvant therapies, triple-negative, and ER+ subtypes of breast cancer (BC) with metastatic potentials remain the leading cause of BC-related deaths. We investigated the role of phosphatidylinositol-4-phosphate 5-kinase alpha (PIP5Kα), a key upstream factor of PI3K/AKT, and the therapeutic effect of PIP5Kα inhibitor on subtypes of BC. The clinical importance of PIP5K1α and its association with survivals were analyzed using three BC cohorts from Nottingham (n = 913), KM plotter (n = 112) and TCGA (n = 817). Targeted overexpression or knockdown of PIP5K1α were introduced into BC cell lines. The effects of PIP5K1α and its inhibitor on growth and invasion of BC were confirmed by using in vitro assays including proliferation, migration, apoptosis and luciferase reporter assays and in vivo xenograft mouse models. All statistical tests were two-sided. PIP5K1α was associated with poor patient outcome in triple-negative BC (for PIP5K1α protein, p = 0.011 and for mRNA expression, p = 0.028, log-rank test). 29% of triple-negative BC had PIP5K1A gene amplification. Elevated level of PIP5K1α increased expression of pSer-473 AKT (p < 0.001) and invasiveness of triple-negative MDA-MB-231 cells (p < 0.001). Conversely, inhibition of PIP5K1α using its inhibitor ISA-2011B, or via knockdown suppressed growth and invasiveness of MDA-MB-231 xenografts (mean vehicle-treated controls = 2160 mm3, and mean ISA-2011B-treated = 600 mm3, p < 0.001). ISA-2011B-treatment reduced expression of pSer-473 AKT (p < 0.001) and its downstream effectors including cyclin D1, VEGF and its receptors, VEGFR1 and VEGFR2 (p < 0.001) in xenograft tumors. In ER+ cancer cells, PIP5K1α acted on pSer-473 AKT, and was in complexes with VEGFR2, serving as co-factor of ER-alpha to regulate activities of target genes including cyclin D1 and CDK1. Our study suggests that our developed PIP5K1α inhibitor has a great potential on refining targeted therapeutics for treatment of triple-negative and ER+ BC with abnormal PI3K/AKT pathways.
Collapse
Affiliation(s)
- Martuza Sarwar
- Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
| | | | - Mohammed Aleskandarany
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Richard Karlsson
- Division of Basal Tumor Biology, Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Maryam Althobiti
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Niels Ødum
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Nigel P Mongan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom.,Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Nisthman Dizeyi
- Division of reproductive research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Heather Johnson
- Department of Bio-Diagnosis, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Andrew R Green
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ian O Ellis
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Emad A Rakha
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jenny L Persson
- Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden. .,Division of Basal Tumor Biology, Department of Molecular Biology, Umeå University, Umeå, Sweden.
| |
Collapse
|
37
|
Kaemmerer E, Turner D, Peters AA, Roberts-Thomson SJ, Monteith GR. An automated epifluorescence microscopy imaging assay for the identification of phospho-AKT level modulators in breast cancer cells. J Pharmacol Toxicol Methods 2018; 92:13-19. [PMID: 29438745 DOI: 10.1016/j.vascn.2018.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/28/2018] [Accepted: 02/08/2018] [Indexed: 11/19/2022]
Abstract
AKT is an enzyme of the PI3K/pAKT pathway, regulating proliferation and cell survival. High basal levels of active, phosphorylated AKT (pAKT) are associated with tumor progression and therapeutic resistance in some breast cancer subtypes, including HER2 positive breast cancers. Various stimuli can increase pAKT levels and elevated basal pAKT levels are a feature of PTEN-deficient breast cancer cell lines. The aim of this study was to develop an assay able to identify modulators of pAKT levels using an automated epifluorescence microscope and high content analysis. To develop this assay, we used HCC-1569, a PTEN-deficient, HER2-overexpressing breast cancer cell line with elevated basal pAKT levels. HCC-1569 cells were treated with a selective pharmacological inhibitor of AKT (MK-2206) to reduce basal pAKT levels or EGF to increase pAKT levels. Immunofluorescence images were acquired using an automated epifluorescence microscope and integrated intensity of cytoplasmic pAKT staining was calculated using high content analysis software. Mean and median integrated cytoplasmic intensity were normalized using fold change and standard score to assess assay quality and to identify most robust data analysis. The highest z' factor was achieved for median data normalization using the standard score method (z' = 0.45). Using our developed assay we identified the calcium homeostasis regulating proteins TPRV6, STIM1 and TRPC1 as modulators of pAKT levels in HCC-1569 cells. Calcium signaling controls a diverse array of cellular processes and some calcium homeostasis regulating proteins are involved in modulating pAKT levels in cancer cells. Thus, these identified hits present promising targets for further assessment.
Collapse
Affiliation(s)
- Elke Kaemmerer
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia.
| | - Dane Turner
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia.
| | - Amelia A Peters
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia.
| | | | - Gregory R Monteith
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
38
|
Moi SH, Lee YC, Chuang LY, Yuan SSF, Ou-Yang F, Hou MF, Yang CH, Chang HW. Cumulative receiver operating characteristics for analyzing interaction between tissue visfatin and clinicopathologic factors in breast cancer progression. Cancer Cell Int 2018; 18:19. [PMID: 29449787 PMCID: PMC5807850 DOI: 10.1186/s12935-018-0517-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 01/31/2018] [Indexed: 01/16/2023] Open
Abstract
Background Visfatin has been reported to be associated with breast cancer progression, but the interaction between the visfatin and clinicopathologic factors in breast cancer progression status requires further investigation. To address this problem, it is better to simultaneously consider multiple factors in sensitivity and specificity assays. Methods In this study, a dataset for 105 breast cancer patients (84 disease-free and 21 progressing) were chosen. Individual and cumulative receiver operating characteristics (ROC) were used to analyze the impact of each factor along with interaction effects. Results In individual ROC analysis, only 3 of 13 factors showed better performance for area under curve (AUC), i.e., AUC > 7 for hormone therapy (HT), tissue visfatin, and lymph node (LN) metastasis. Under our proposed scoring system, the cumulative ROC analysis provides higher AUC performance (0.746–0.886) than individual ROC analysis in predicting breast cancer progression. Considering the interaction between these factors, a minimum of six factors, including HT, tissue visfatin, LN metastasis, tumor stage, age, and tumor size, were identified as being highly interactive and associated with breast cancer progression, providing potential and optimal discriminators for predicting breast cancer progression. Conclusion Taken together, the cumulative ROC analysis provides better prediction for breast cancer progression than individual ROC analysis. Electronic supplementary material The online version of this article (10.1186/s12935-018-0517-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sin-Hua Moi
- 1Department of Electronic Engineering, National Kaohsiung University of Applied Sciences, Kaohsiung, Taiwan
| | - Yi-Chen Lee
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,3Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Yeh Chuang
- 4Department of Chemical Engineering & Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung, Taiwan
| | - Shyng-Shiou F Yuan
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fu Ou-Yang
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,6Division of Breast Surgery and Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ming-Feng Hou
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,7Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,8Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Hong Yang
- 1Department of Electronic Engineering, National Kaohsiung University of Applied Sciences, Kaohsiung, Taiwan.,9Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsueh-Wei Chang
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,10Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan.,11Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,12Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
39
|
Peng F, Tang H, Liu P, Shen J, Guan X, Xie X, Gao J, Xiong L, Jia L, Chen J, Peng C. Isoliquiritigenin modulates miR-374a/PTEN/Akt axis to suppress breast cancer tumorigenesis and metastasis. Sci Rep 2017; 7:9022. [PMID: 28827662 PMCID: PMC5567123 DOI: 10.1038/s41598-017-08422-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/10/2017] [Indexed: 01/15/2023] Open
Abstract
Breast cancer is one of the most frightful causes of death among females worldwide. Accumulating evidence attached the importance of microRNAs negative regulation to tumorigenesis in breast cancer, suggesting novel cancer therapies targeting microRNAs modulation. Recent studies demonstrated that isoliquiritigenin could inhibit breast cancer cells proliferation and migration, but the underlying mechanism is still limited. In this study, the anti-cancer effects as well as the detailed mechanisms of isoliquiritigenin were explored. The results proved that isoliquiritigenin could negatively regulate breast cancer growth through the induction of apoptosis. We also verified the anti-cancer effect of isoliquiritigenin on migration and invasion, and identified highly expressed miR-374a as one of the main microRNAs down-regulated by isoliquiritigenin treatment in breast cancer. Further study displayed that isoliquiritigenin increased PTEN expression through the decrease of miR-374a expression to inhibit the aberrant Akt signaling. Our findings suggest isoliquiritigenin as a novel anti-cancer candidate significantly regulating miR-374a/PTEN/Akt axis in microRNA-based breast cancer therapies.
Collapse
Affiliation(s)
- Fu Peng
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Chengdu University of Traditional Chinese Medicine, Chengdu, China.,State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Sichuan Province and Ministry of Science and Technology, Chengdu, China
| | - Hailin Tang
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Breast Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Peng Liu
- Department of Breast Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xinyuan Guan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Xiaofang Xie
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.,State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Sichuan Province and Ministry of Science and Technology, Chengdu, China
| | - Jihai Gao
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.,State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Sichuan Province and Ministry of Science and Technology, Chengdu, China
| | - Liang Xiong
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.,State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Sichuan Province and Ministry of Science and Technology, Chengdu, China
| | - Lei Jia
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong. .,Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Cheng Peng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China. .,State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Sichuan Province and Ministry of Science and Technology, Chengdu, China.
| |
Collapse
|
40
|
Overexpression of phosphoprotein phosphatase 2A predicts worse prognosis in patients with breast cancer: a 15-year follow-up. Hum Pathol 2017; 66:93-100. [PMID: 28603063 DOI: 10.1016/j.humpath.2017.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/26/2017] [Accepted: 06/01/2017] [Indexed: 12/15/2022]
Abstract
Breast cancer subtypes can be stratified by IHC expression of estrogen receptor, progesterone receptor, and human epidermal growth factor 2 (HER2). The signaling pathways mediated by these receptors are the dominant drivers of cell proliferation and survival in most human breast cancers. One of the most frequently overactivated pathways in breast cancer is the AKT signaling cascade. Protein phosphatase 2A (PP2A) acts as a switch to turn off signal transduction in the AKT pathway; however, it is frequently inactivated in many cancers by phosphorylation of Tyr-307 to form phosphoprotein phosphatase 2A (p-PP2A). This study aimed to investigate the clinical significance of p-PP2A and phospho-AKT (p-AKT) expression in 672 patients with breast cancer during a 15-year follow-up. The breast tissue microarray was evaluated for p-PP2A and p-AKT expression using IHC staining and scores. Analysis of IHC staining results revealed that p-PP2A expression was positively correlated with HER2, Ki-67, and p-AKT overexpression (P<.001, P=.003, and P=.001, respectively). At the time of diagnosis, breast cancer patients with higher p-PP2A expression had significantly shorter 15-year OS than patients with lower p-PP2A expression did (P=.017). Multivariate Cox regression analysis revealed that high p-PP2A expression was an independent prognostic factor for shorter OS (hazard ratio, 1.741; P=.012). Our data revealed that high p-PP2A expression is positively associated with HER2, Ki-67, and p-AKT expression. High p-PP2A expression correlates with poor clinical outcomes in breast cancer, especially in patients with TNBC.
Collapse
|
41
|
Wu Y, Zhang D, Wu B, Quan Y, Liu D, Li Y, Zhang X. Synergistic Activity of an Antimetabolite Drug and Tyrosine Kinase Inhibitors against Breast Cancer Cells. Chem Pharm Bull (Tokyo) 2017; 65:768-775. [PMID: 28539531 DOI: 10.1248/cpb.c17-00261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Antimetabolite drugs, including the adenosine deaminase inhibitor cladribine, have been shown to induce apoptosis in a variety of cancer cells, and have been widely used in clinical trials of various cancers in conjunction with tyrosine kinase inhibitors (TKIs). Combination treatment with cladribine and gefitinib or dasatinib is expected to have a synergistic inhibitory effect on breast cancer cell growth. Our results demonstrated that the combination treatment had synergistic activity against human breast cancer (MCF-7) cells, enhanced G2/M cell arrest and reactive oxygen species (ROS) generation, and increased the loss of mitochondrial membrane potential and cell apoptosis. In addition, the combination treatment decreased Bcl-2 expression. Our results demonstrated that cladribine in combination with gefitinib or dasatinib exerted synergistic anticancer effects on MCF-7 cells by inducing cell cycle arrest, ROS production and apoptosis through the mitochondria-mediated intrinsic pathway.
Collapse
Affiliation(s)
- Yushan Wu
- School of Life Sciences, Shandong University of Technology
| | - Dongxing Zhang
- School of Life Sciences, Shandong University of Technology
| | - Baofan Wu
- Medical Department, Heze Municipal Hospital
| | - Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University
| | - Dongwu Liu
- School of Life Sciences, Shandong University of Technology
| | | | - Xiuzhen Zhang
- School of Life Sciences, Shandong University of Technology
| |
Collapse
|
42
|
Ma M, Ma Y, Zhang GJ, Liao R, Jiang XF, Yan XX, Bie FJ, Li XB, Lv YH. Eugenol alleviated breast precancerous lesions through HER2/PI3K-AKT pathway-induced cell apoptosis and S-phase arrest. Oncotarget 2017; 8:56296-56310. [PMID: 28915591 PMCID: PMC5593562 DOI: 10.18632/oncotarget.17626] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 04/19/2017] [Indexed: 01/19/2023] Open
Abstract
Eugenol can be separated from the oil extract of clove bud, and has many pharmacological functions such as anticancer and transdermal absorption. HER2/PI3K-AKT is a key signaling pathway in the development of breast cancer. In this study, 80 μM eugenol could significantly inhibit the proliferation of HER-2 positive MCF-10AT cells and the inhibition rate was up to 32.8%, but had no obvious inhibitory effect on MCF-7 and MCF-10A cells with HER2 weak expression. Eugenol also significantly induced human breast precancerous lesion MCF-10AT cell apoptosis and cell cycle S-phase arrest, but the biological effects nearly disappeared after HER2 over-expression through transfecting pcDNA3.1-HER2. In MCF-10AT cells treated by 180 μM eugenol, the protein expressions of HER2, AKT, PDK1, p85, Bcl2, NF-κB, Bad and Cyclin D1 were decreased and the decreased rates were respectively 63.0%, 60.0%, 52.9%, 62.9%, 37.1%, 47.2%, 61.7%, 59.1%, while the p21, p27 and Bax expression were increased by 4.48-, 4.76- and 2.57-fold respectively. In the rat models of breast precancerous lesion, 1 mg eugenol for external use significantly inhibited the progress of breast precancerous lesion and the occurrence rate of breast precancerous lesions and invasive carcinomas was decreased by about 30.5%. Furthermore eugenol for external (1 mg) markedly decreased the protein expressions of HER2 (62.9%), AKT (58.6%), PDK1 (56.4%), p85 (54.3%), Bcl2 (59.3%), NF-κB (65.7%), Bad (64.0%), Cyclin D1 (43.0%), while p21, p27 and Bax protein expressions were respectively increased 1.83-, 2.52- and 2.51-fold. The results showed eugenol could significantly inhibit the development of breast precancerous lesions by blocking HER2/PI3K-AKT signaling network. So eugenol may be a promising external drug for breast precancerous lesions.
Collapse
Affiliation(s)
- Min Ma
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Yi Ma
- Institute of Biomedicine, Department of Cellular Biology, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Gui-Juan Zhang
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Rui Liao
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Xue-Feng Jiang
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Xian-Xin Yan
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Feng-Jie Bie
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Xiao-Bo Li
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Yan-Hong Lv
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| |
Collapse
|
43
|
Cao F, Zhang C, Han W, Gao XJ, Ma J, Hu YW, Gu X, Ding HZ, Zhu LX, Liu Q. p-Akt as a potential poor prognostic factor for gastric cancer: a systematic review and meta-analysis. Oncotarget 2017; 8:59878-59888. [PMID: 28938690 PMCID: PMC5601786 DOI: 10.18632/oncotarget.17001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/17/2017] [Indexed: 12/18/2022] Open
Abstract
To understand the relationship between p-Akt expression and the prognosis of patients with gastric cancer, we searched six databases, Pubmed, EMBASE, Cochrane Library, CNKI, Wanfang and CBM for relevant articles in order to conduct this metaanalysis. The pooled hazard ratios and corresponding 95%CI of overall survival were calculated to evaluate the prognostic value of p-Akt expression in patients with gastric cancer. With 2261 patients combined from 13 available studies, the pooled HR showed a poor prognosis in patients with gastric cancer in the univariate analysis (HR=1.88, 95%CI:1.45-2.43, P<0.00001), and the group "univariate analysis+estimate" (HR=1.41, 95%CI: 1.01-1.97, P=0.04), but not in multivariate analysis (HR=0.66, 95%CI: 0.29-1.52, P=0.33) and estimate (HR=1.13, 95%CI: 0.65-1.95, P=0.67). In conclusion, our results indicated that p-Akt was likely to be an indicator of poor prognosis in patients with gastric cancer.
Collapse
Affiliation(s)
- Fang Cao
- Department of General Surgery, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Cong Zhang
- Department of Pharmacy, Kunshan Hospital of TCM, Kunshan, Jiangsu 215300, P.R. China
| | - Wei Han
- Department of General Surgery, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Xiao-Jiao Gao
- Department of Pathology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Jun Ma
- Department of Urological Surgery, Kunshan Hospital of TCM, Kunshan, Jiangsu 215300, P.R. China
| | - Yong-Wei Hu
- Department of General Surgery, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Xing Gu
- Department of Gynaecology and Obstetrics, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Hou-Zhong Ding
- Department of General Surgery, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Li-Xia Zhu
- Department of Gynaecology and Obstetrics, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Qin Liu
- Department of Gynaecology and Obstetrics, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| |
Collapse
|
44
|
Interaction of MRE11 and Clinicopathologic Characteristics in Recurrence of Breast Cancer: Individual and Cumulated Receiver Operating Characteristic Analyses. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2563910. [PMID: 28133604 PMCID: PMC5241446 DOI: 10.1155/2017/2563910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/28/2016] [Indexed: 12/28/2022]
Abstract
The interaction between the meiotic recombination 11 homolog A (MRE11) oncoprotein and breast cancer recurrence status remains unclear. The aim of this study was to assess the interaction between MRE11 and clinicopathologic variables in breast cancer. A dataset for 254 subjects with breast cancer (220 nonrecurrent and 34 recurrent) was used in individual and cumulated receiver operating characteristic (ROC) analyses of MRE11 and 12 clinicopathologic variables for predicting breast cancer recurrence. In individual ROC analysis, the area under curve (AUC) for each predictor of breast cancer recurrence was smaller than 0.7. In cumulated ROC analysis, however, the AUC value for each predictor improved. Ten relevant variables in breast cancer recurrence were used to find the optimal prognostic indicators. The presence of any six of the following ten variables had a high (79%) sensitivity and a high (70%) specificity for predicting breast cancer recurrence: tumor size ≥ 2.4 cm, tumor stage II/III, therapy other than hormone therapy, age ≥ 52 years, MRE11 positive cells > 50%, body mass index ≥ 24, lymph node metastasis, positivity for progesterone receptor, positivity for epidermal growth factor receptor, and negativity for estrogen receptor. In conclusion, this study revealed that these 10 clinicopathologic variables are the minimum discriminators needed for optimal discriminant effectiveness in predicting breast cancer recurrence.
Collapse
|
45
|
Chen Q, Weng Z, Lu Y, Jia Y, Ding L, Bai F, Ge M, Lin Q, Wu K. An Experimental Analysis of the Molecular Effects of Trastuzumab (Herceptin) and Fulvestrant (Falsodex), as Single Agents or in Combination, on Human HR+/HER2+ Breast Cancer Cell Lines and Mouse Tumor Xenografts. PLoS One 2017; 12:e0168960. [PMID: 28045951 PMCID: PMC5207527 DOI: 10.1371/journal.pone.0168960] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/08/2016] [Indexed: 11/28/2022] Open
Abstract
PURPOSE To investigate the effects of trastuzumab (herceptin) and fulvestrant (falsodex) either in combination or alone, on downstream cell signaling pathways in lab-cultured human HR+/HER2+ breast cancer cell lines ZR-75-1 and BT-474, as well as on protein expression levels in mouse xenograft tissue. METHODS Cells were cultivated in the presence of trastuzumab or fulvestrant or both. Molecular events that resulted in an inhibition of cell proliferation and cell cycle progression or in an increased rate of apoptosis were studied. The distribution and abundance of the proteins p-Akt and p-Erk expressed in these cells in response to single agents or combinatorial treatment were also investigated. In addition, the effects of trastuzumab and fulvestrant, either as single agents or in combination on tumor growth as well as on expression of the protein p-MED1 expressed in in vivo mouse xenograft models was also examined. RESULTS Cell proliferation was increasingly inhibited by trastuzumab or fulvestrant or both, with a CI<1 and DRI>1 in both human cell lines. The rate of apoptosis increased only in the BT-474 cell line and not in the ZR-75-1 cell line upon treatment with fulvestrant and not trastuzumab as a single agent (P<0.05). Interestingly, fulvestrant, in combination with trastuzumab, did not significantly alter the rate of apoptosis (in comparison with fulvestrant alone), in the BT-474 cell line (P>0.05). Cell accumulation in the G1 phase of cell cycle was investigated in all treatment groups (P<0.05), and the combination of trastuzumab and fulvestrant reversed the effects of fulvestrant alone on p-Akt and p-Erk protein expression levels. Using ZR-75-1 or BT-474 to generate in vivo tumor xenografts in BALB/c athymic mouse models, we showed that a combination of both drugs resulted in a stronger inhibition of tumor growth (P<0.05) and a greater decrease in the levels of activated MED1 (p-MED1) expressed in tumor issues compared with the use of either drug as a single agent. CONCLUSIONS We demonstrate that the administration of trastuzumab and fulvestrant in combination results in positive synergistic effects on both, ZR-75-1 and BT-474 cell lines. This combinatorial approach is likely to reduce physiological side effects of both drugs, thus providing a theoretical basis for the use of such combination treatment in order to resolve HR+/HER2+ triple positive breast cancer that has previously been shown to be resistant to endocrine treatment alone.
Collapse
Affiliation(s)
- Qing Chen
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyi Weng
- Department of General Surgery, Shanghai International Medical Center, Shanghai, China
| | - Yunshu Lu
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yijun Jia
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Longlong Ding
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Bai
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meixin Ge
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Lin
- Department of Radiation Oncology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Kejin Wu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
46
|
Estrogen Enhances the Cell Viability and Motility of Breast Cancer Cells through the ERα-ΔNp63-Integrin β4 Signaling Pathway. PLoS One 2016; 11:e0148301. [PMID: 26845172 PMCID: PMC4742232 DOI: 10.1371/journal.pone.0148301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 01/15/2016] [Indexed: 12/19/2022] Open
Abstract
Estrogen induces ERα-positive breast cancer aggressiveness via the promotion of cell proliferation and survival, the epithelial-mesenchymal transition, and stem-like properties. Integrin β4 signaling has been implicated in estrogen/ERα-induced tumorigenicity and anti-apoptosis; however, this signaling cascade poorly understood. ΔNp63, an N-terminally truncated isoform of the p63 transcription factor, functions as a transcription factor of integrinβ4 and therefore regulates cellular adhesion and survival. Therefore, the aim of the present study was to investigate the estrogen-induced interaction between ERα, ΔNp63 and integrin β4 in breast cancer cells. In ERα-positive MCF-7 cells, estrogen activated ERα transcription, which induced ΔNp63 expression. And ΔNp63 subsequently induced integrin β4 expression, which resulted in AKT phosphorylation and enhanced cell viability and motility. Conversely, there was no inductive effect of estrogen on ΔNp63-integrinβ4-AKT signaling or on cell viability and motility in ERα-negative MDA-MB-231 cells. ΔNp63 knockdown abolishes these estrogen-induced effects and reduces cell viability and motility in MCF-7 cells. Nevertheless, ΔNp63 knockdown also inhibited cell migration in MDA-MB-231 cells through reducing integrin β4 expression and AKT phosphorylation. In conclusion, estrogen enhances ERα-positive breast cancer cell viability and motility through activating the ERα-ΔNp63-integrin β4 signaling pathway to induce AKT phosphorylated activation. Those findings should be useful to elucidate the crosstalk between estrogen/ER signaling and ΔNp63 signaling and provide novel insights into the effects of estrogen on breast cancer progression.
Collapse
|
47
|
Robbins HL, Hague A. The PI3K/Akt Pathway in Tumors of Endocrine Tissues. Front Endocrinol (Lausanne) 2015; 6:188. [PMID: 26793165 PMCID: PMC4707207 DOI: 10.3389/fendo.2015.00188] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/07/2015] [Indexed: 12/29/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is a key driver in carcinogenesis. Defects in this pathway in human cancer syndromes such as Cowden's disease and Multiple Endocrine Neoplasia result in tumors of endocrine tissues, highlighting its importance in these cancer types. This review explores the growing evidence from multiple animal and in vitro models and from analysis of human tumors for the involvement of this pathway in the following: thyroid carcinoma subtypes, parathyroid carcinoma, pituitary tumors, adrenocortical carcinoma, phaeochromocytoma, neuroblastoma, and gastroenteropancreatic neuroendocrine tumors. While data are not always consistent, immunohistochemistry performed on human tumor tissue has been used alongside other techniques to demonstrate Akt overactivation. We review active Akt as a potential prognostic marker and the PI3K pathway as a therapeutic target in endocrine neoplasia.
Collapse
Affiliation(s)
- Helen Louise Robbins
- Department of General Surgery, University Hospital Coventry and Warwickshire, Coventry, UK
| | - Angela Hague
- School of Oral and Dental Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
- *Correspondence: Angela Hague,
| |
Collapse
|