1
|
Yang Y, Zhang S, Su S, Yang X, Chen J, Sang A. The Effects of STRA6 Regulation of the Circadian Rhythm on Choroidal Neovascularization. Invest Ophthalmol Vis Sci 2024; 65:21. [PMID: 39269368 PMCID: PMC11407478 DOI: 10.1167/iovs.65.11.21] [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: 04/16/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Purpose This study aims to investigate the relationship among STRA6, circadian rhythm, and choroidal neovascularization (CNV) formation, as well as the regulatory mechanism of STRA6 in CNV under circadian rhythm disturbances. Methods C57BL/6J male mice (aged 6 weeks) were randomly divided into control and jet lag groups (using a time shift method every 4 days to disrupt the molecular clock's capacity to synchronize with a stable rhythm). A laser-induced CNV model was established in both the control and the jet lag group after 2 weeks of jet lag. The size of CNV lesions and vascular leakage were detected by morphological and imaging examination on the seventh day post laser. STRA6 was screened by full transcriptome sequencing. Bioinformatics analysis was conducted to assess the variation and association of STRA6 in the GSE29801 dataset. The effects of STRA6 were evaluated both in vivo and in vitro. The pathway mechanism was further elucidated and confirmed through immunofluorescence of paraffin sections and Western blotting. Results The disturbance of circadian rhythm promotes the formation of CNV. Patients with age-related macular degeneration (AMD) exhibited higher levels of STRA6 expression compared to the control group, and STRA6 was enriched in pathways related to angiogenesis. In addition, CLOCK and BMAL1, which are initiators that drive the circadian cycle, had regulatory effects on STRA6. Knocking down STRA6 reversed the promotion of CNV formation caused by circadian rhythm disturbance in vivo, and it also affected the proliferation, migration, and VEGF secretion of RPE cells without circadian rhythm in vitro, as well as impacting endothelial cells. Through activation of the JAK2/STAT3/VEGFA signaling pathway in unsynchronized RPE cells, STRA6 promotes CNV formation. Conclusions This study suggests that STRA6 reduces CNV production by inhibiting JAK2/STAT3 phosphorylation after circadian rhythm disturbance. The results suggest that STRA6 may be a new direction for the treatment of AMD.
Collapse
Affiliation(s)
- Ying Yang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Shenglai Zhang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Shu Su
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiaowei Yang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jia Chen
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Aimin Sang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
2
|
Jia J, He R, Yao Z, Su J, Deng S, Chen K, Yu B. Daidzein alleviates osteoporosis by promoting osteogenesis and angiogenesis coupling. PeerJ 2023; 11:e16121. [PMID: 37868048 PMCID: PMC10586307 DOI: 10.7717/peerj.16121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/27/2023] [Indexed: 10/24/2023] Open
Abstract
Background Postmenopausal osteoporosis and osteoporosis-related fractures are world-wide serious public health problem. Recent studies demonstrated that inhibiting caveolin-1 leads to osteoclastogenesis suppression and protection against OVX-induced osteoporosis. This study aimed to explore the mechanism of caveolin-1 mediating bone loss and the potential therapeutic target. Methods Thirty C57BL/6 female mice were allocated randomly into three groups: sham or bilateral ovariectomy (OVX) surgeries were performed for mice and subsequently daidzein or vehicle was administrated to animals (control, OVX + vehicle and OVX + daidzein). After 8-week administration, femurs were harvested for Micro-CT scan, histological staining including H&E, immunohistochemistry, immunofluorescence, TRAP. Bone marrow endothelial cells (BMECs) were cultured and treated with inhibitors of caveolin-1 (daidzein) or EGFR (erlotinib) and then scratch wound healing and ki67 assays were performed. In addition, cells were harvested for western blot and PCR analysis. Results Micro-CT showed inhibiting caveolin-1with daidzein alleviated OVX-induced osteoporosis and osteogenesis suppression. Further investigations revealed H-type vessels in cancellous bone were decreased in OVX-induced mice, which can be alleviated by daidzein. It was subsequently proved that daidzein improved migration and proliferation of BMECs hence improved H-type vessels formation through inhibiting caveolin-1, which suppressed EGFR/AKT/PI3K signaling in BMECs. Conclusions This study demonstrated that daidzein alleviates OVX-induced osteoporosis by promoting H-type vessels formation in cancellous bone, which then promotes bone formation. Activating EGFR/AKT/PI3K signaling could be the critical reason.
Collapse
Affiliation(s)
- Junjie Jia
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopaedics, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Ruiyi He
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopaedics, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Zilong Yao
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianwen Su
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Songyun Deng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kun Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Dilworth L, Stennett D, Omoruyi F. Cellular and Molecular Activities of IP6 in Disease Prevention and Therapy. Biomolecules 2023; 13:972. [PMID: 37371552 DOI: 10.3390/biom13060972] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
IP6 (phytic acid) is a naturally occurring compound in plant seeds and grains. It is a poly-phosphorylated inositol derivative that has been shown to exhibit many biological activities that accrue benefits in health and diseases (cancer, diabetes, renal lithiasis, cardiovascular diseases, etc.). IP6 has been shown to have several cellular and molecular activities associated with its potential role in disease prevention. These activities include anti-oxidant properties, chelation of metal ions, inhibition of inflammation, modulation of cell signaling pathways, and modulation of the activities of enzymes and hormones that are involved in carbohydrate and lipid metabolism. Studies have shown that IP6 has anti-oxidant properties and can scavenge free radicals known to cause cellular damage and contribute to the development of chronic diseases such as cancers and cardiovascular diseases, as well as diabetes mellitus. It has also been shown to possess anti-inflammatory properties that may modulate immune responses geared towards the prevention of inflammatory conditions. Moreover, IP6 exhibits anti-cancer properties through the induction of cell cycle arrest, promoting apoptosis and inhibiting cancer cell growth. Additionally, it has been shown to have anti-mutagenic properties, which reduce the risk of malignancies by preventing DNA damage and mutations. IP6 has also been reported to have a potential role in bone health. It inhibits bone resorption and promotes bone formation, which may help in the prevention of bone diseases such as osteoporosis. Overall, IP6's cellular and molecular activities make it a promising candidate for disease prevention. As reported in many studies, its anti-inflammatory, anti-oxidant, and anti-cancer properties support its inclusion as a dietary supplement that may protect against the development of chronic diseases. However, further studies are needed to understand the mechanisms of action of this dynamic molecule and its derivatives and determine the optimal doses and appropriate delivery methods for effective therapeutic use.
Collapse
Affiliation(s)
- Lowell Dilworth
- Department of Pathology, The University of the West Indies, Mona Campus, Kingston 7, Jamaica
| | - Dewayne Stennett
- The Transitional Year Programme, University of Toronto, Toronto, ON M5S 2E8, Canada
| | - Felix Omoruyi
- Department of Life Sciences, Texas A&M University, Corpus Christi, TX 78412, USA
- Department of Health Sciences, Texas A&M University, Corpus Christi, TX 78412, USA
| |
Collapse
|
4
|
Xu H, Wang W, Liu X, Huang W, Zhu C, Xu Y, Yang H, Bai J, Geng D. Targeting strategies for bone diseases: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:202. [PMID: 37198232 DOI: 10.1038/s41392-023-01467-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/02/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023] Open
Abstract
Since the proposal of Paul Ehrlich's magic bullet concept over 100 years ago, tremendous advances have occurred in targeted therapy. From the initial selective antibody, antitoxin to targeted drug delivery that emerged in the past decades, more precise therapeutic efficacy is realized in specific pathological sites of clinical diseases. As a highly pyknotic mineralized tissue with lessened blood flow, bone is characterized by a complex remodeling and homeostatic regulation mechanism, which makes drug therapy for skeletal diseases more challenging than other tissues. Bone-targeted therapy has been considered a promising therapeutic approach for handling such drawbacks. With the deepening understanding of bone biology, improvements in some established bone-targeted drugs and novel therapeutic targets for drugs and deliveries have emerged on the horizon. In this review, we provide a panoramic summary of recent advances in therapeutic strategies based on bone targeting. We highlight targeting strategies based on bone structure and remodeling biology. For bone-targeted therapeutic agents, in addition to improvements of the classic denosumab, romosozumab, and PTH1R ligands, potential regulation of the remodeling process targeting other key membrane expressions, cellular crosstalk, and gene expression, of all bone cells has been exploited. For bone-targeted drug delivery, different delivery strategies targeting bone matrix, bone marrow, and specific bone cells are summarized with a comparison between different targeting ligands. Ultimately, this review will summarize recent advances in the clinical translation of bone-targeted therapies and provide a perspective on the challenges for the application of bone-targeted therapy in the clinic and future trends in this area.
Collapse
Affiliation(s)
- Hao Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wentao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Xin Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Chen Zhu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
5
|
Sánchez-García J, Muñoz-Pina S, García-Hernández J, Heredia A, Andrés A. Impact of Air-Drying Temperature on Antioxidant Properties and ACE-Inhibiting Activity of Fungal Fermented Lentil Flour. Foods 2023; 12:999. [PMID: 36900516 PMCID: PMC10001291 DOI: 10.3390/foods12050999] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Solid-state fermentation (SSF) with Pleurotus ostreatus enhances the nutritional value of legumes. However, drying can cause significant changes in physical and nutritional properties of the final products. Thus, this work studies the impact of air-drying temperature (50, 60, and 70 °C) on relevant properties (antioxidant properties, ACE-inhibitory capacity, phytic acid, colour, and particle size) of two fermented lentils flour (Pardina and Castellana) using freeze-drying as a reference method. Castellana variety is a better substrate for Pleurotus, generating four times more biomass. In addition, an almost total reduction of phytic acid from 7.3 to 0.9 mg/g db is achieved in this variety. Air-drying significantly decreased the particle size and the final colour with ΔE > 20; nonetheless, the temperature does not play a crucial role. SSF decreased the total phenolic content and the antioxidant capacity regardless of the variety, however, drying at 70 °C increased total phenolic content (186%) in fermented Castellana flour. Comparing drying methods, freeze-drying implied a higher decrease in those parameters, reducing the TPC from 2.4 to 1.6 and from 7.7 to 3.4 mg gallic acid/g db in Pardina and Castellana dried flours. Finally, the flours inhibit the angiotensin I-converting-enzyme, and fermentation and drying increased their potential cardiovascular benefits.
Collapse
Affiliation(s)
- Janaina Sánchez-García
- Instituto Universitario de Ingeniería de Alimentos para el Desarrollo (IUIAD-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| | - Sara Muñoz-Pina
- Instituto Universitario de Ingeniería de Alimentos para el Desarrollo (IUIAD-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| | - Jorge García-Hernández
- Centro Avanzado de Microbiología de Alimentos (CAMA), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| | - Ana Heredia
- Instituto Universitario de Ingeniería de Alimentos para el Desarrollo (IUIAD-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| | - Ana Andrés
- Instituto Universitario de Ingeniería de Alimentos para el Desarrollo (IUIAD-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| |
Collapse
|
6
|
Raina K, Kandhari K, Jain AK, Ravichandran K, Maroni P, Agarwal C, Agarwal R. Stage-Specific Effect of Inositol Hexaphosphate on Cancer Stem Cell Pool during Growth and Progression of Prostate Tumorigenesis in TRAMP Model. Cancers (Basel) 2022; 14:4204. [PMID: 36077751 PMCID: PMC9455012 DOI: 10.3390/cancers14174204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/23/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Herein, we assessed the stage-specific efficacy of inositol hexaphosphate (IP6, phytic acid), a bioactive food component, on prostate cancer (PCa) growth and progression in a transgenic mouse model of prostate cancer (TRAMP). Starting at 4, 12, 20, and 30 weeks of age, male TRAMP mice were fed either regular drinking water or 2% IP6 in water for ~8-15 weeks. Pathological assessments at study endpoint indicated that tumor grade is arrested at earlier stages by IP6 treatment; IP6 also prevented progression to more advanced forms of the disease (~55-70% decrease in moderately and poorly differentiated adenocarcinoma incidence was observed in advanced stage TRAMP cohorts). Next, we determined whether the protective effects of IP6 are mediated via its effect on the expansion of the cancer stem cells (CSCs) pool; results indicated that the anti-PCa effects of IP6 are associated with its potential to eradicate the PCa CSC pool in TRAMP prostate tumors. Furthermore, in vitro assays corroborated the above findings as IP6 decreased the % of floating PC-3 prostaspheres (self-renewal of CSCs) by ~90%. Together, these findings suggest the multifaceted chemopreventive-translational potential of IP6 intervention in suppressing the growth and progression of PCa and controlling this malignancy at an early stage.
Collapse
Affiliation(s)
- Komal Raina
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Kushal Kandhari
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anil K. Jain
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kameswaran Ravichandran
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paul Maroni
- Department of Surgery, Division of Urology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
7
|
Salazar AS, Rakhmankulova M, Simon LE, Toriola AT. Chemoprevention Agents to Reduce Mammographic Breast Density in Premenopausal Women: A Systematic Review of Clinical Trials. JNCI Cancer Spectr 2021; 5:pkaa125. [PMID: 33554041 PMCID: PMC7853173 DOI: 10.1093/jncics/pkaa125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/05/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Background Higher mammographic breast density (MBD) is associated with an increased risk of breast cancer when compared with lower MBD, especially in premenopausal women. However, little is known about the effectiveness of chemoprevention agents in reducing MBD in premenopausal women without a history of breast cancer. Findings from this review should provide insight on how to target MBD in breast cancer prevention in premenopausal women with dense breasts. Methods We searched 9 electronic databases for clinical trials in English, Spanish, French, or German published until January 2020. Articles evaluating the association of pharmacological agents and MBD were included. Data were extracted on methods, type and dose of intervention, outcomes, side effects, and follow up. Quality of the studies was assessed using the US Preventive Services Task Force criteria. Results We identified 7 clinical trials evaluating the associations of 6 chemoprevention agents with changes in MBD in premenopausal women without history of breast cancer. The studies evaluated selective estrogen-receptor modulators (n = 1); gonadotropin-releasing hormone agonists (n = 2); isoflavones (n = 1); vitamin D (n = 1); and Boswellia, betaine, and mayo-inositol compound (n = 1). Hormonal interventions were associated with net reductions in percent density (tamoxifen [13.4%], leuprolide acetate [8.9%], and goserelin [2.7%]), whereas nonhormonal (vitamin D and isoflavone) interventions were not. However, MBD returned to preintervention baseline levels after cessation of gonadotropin-releasing hormone agonists. Conclusions A limited number of chemoprevention agents have been shown to reduce MBD in premenopausal women. Identification of new and well-tolerated chemoprevention agents targeting MBD and larger studies to confirm agents that have been studied in small trials are urgent priorities for primary breast cancer prevention in premenopausal women with dense breasts.
Collapse
Affiliation(s)
- Ana S Salazar
- Department of Surgery, Division of Public Health Sciences, Washington University School of Medicine, St Louis, MO, USA
| | - Malika Rakhmankulova
- Department of Surgery, Division of Public Health Sciences, Washington University School of Medicine, St Louis, MO, USA
| | - Laura E Simon
- Bernard Becker Medical Library, Washington University School of Medicine, St Louis, MO, USA
| | - Adetunji T Toriola
- Department of Surgery, Division of Public Health Sciences, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
8
|
Grapefruit-Derived Micro and Nanovesicles Show Distinct Metabolome Profiles and Anticancer Activities in the A375 Human Melanoma Cell Line. Cells 2020; 9:cells9122722. [PMID: 33371199 PMCID: PMC7766354 DOI: 10.3390/cells9122722] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Fruit juice is one of the most easily accessible resources for the isolation of plant-derived vesicles. Here we found that micro- and nano-sized vesicles (MVs and NVs) from four Citrus species, C. sinensis, C. limon, C. paradisi and C. aurantium, specifically inhibit the proliferation of lung, skin and breast cancer cells, with no substantial effect on the growth of non-cancer cells. Cellular and molecular analyses demonstrate that grapefruit-derived vesicles cause cell cycle arrest at G2/M checkpoint associated with a reduced cyclins B1 and B2 expression levels and the upregulation of cell cycle inhibitor p21. Further data suggest the inhibition of Akt and ERK signalling, reduced intercellular cell adhesion molecule-1 and cathepsins expressions, and the presence of cleaved PARP-1, all associated with the observed changes at the cellular level. Gas chromatography-mass spectrometry-based metabolomics reveals distinct metabolite profiles for the juice and vesicle fractions. NVs exhibit a high relative amount of amino acids and organic acids whereas MVs and fruit juice are characterized by a high percentage of sugars and sugar derivatives. Grapefruit-derived NVs are in particular rich in alpha–hydroxy acids and leucine/isoleucine, myo-inositol and doconexent, while quininic acid was detected in MVs. Our findings reveal the metabolite signatures of grapefruit-derived vesicles and substantiate their potential use in new anticancer strategies.
Collapse
|
9
|
Wang C, Li L, Zhang S, Yan Y, Huang Q, Cai X, Xiao J, Cheng Y. Carrier-Free Platinum Nanomedicine for Targeted Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004829. [PMID: 33205610 DOI: 10.1002/smll.202004829] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/11/2020] [Indexed: 05/06/2023]
Abstract
Numerous nanomedicines have been developed to improve the efficiency and safety of conventional anticancer drugs; however, the complexities in carrier materials and functional integration make it challenging to promote these candidates for clinical translation. In this study, a facile method to prepare carrier-free anticancer nanodrug with inherent bone targeting and osteoclastogenesis inhibition capabilities is reported. Phytic acid, a naturally occurring and nontoxic product, is reacted with cisplatin to form uniform nanoparticles of different sizes. The prepared nanoparticles possess high drug loading and pH-responsive drug release behaviors. Phytic acid in the nanomedicine ensures high bone targeting and osteoclastogenesis inhibition, and the released platinum drugs triggered by tumor extracellular acidity eradicate tumor cells. The nanomedicine around 100 nm shows high anticancer activity and much reduced side effects in a subcutaneous breast cancer model when compared with cisplatin. In addition, it shows high accumulation at osteolytic lesions, and efficiently inhibits tumor growth and tumor-associated osteolysis in a bone metastatic breast cancer model. Here, a facile and efficient strategy to prepare carrier-free nanomedicines with high anticancer drug loading, inherent bone targeting, and osteoclast inhibitory activities for cancer therapy is provided.
Collapse
Affiliation(s)
- Changping Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
| | - Lin Li
- Department of Orthopedics Oncology, Changzheng Hospital, The Second Military Medical University, Shanghai, 200003, P. R. China
| | - Song Zhang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
| | - Yang Yan
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Quan Huang
- Department of Orthopedics Oncology, Changzheng Hospital, The Second Military Medical University, Shanghai, 200003, P. R. China
| | - Xiaopan Cai
- Department of Orthopedics Oncology, Changzheng Hospital, The Second Military Medical University, Shanghai, 200003, P. R. China
| | - Jianru Xiao
- Department of Orthopedics Oncology, Changzheng Hospital, The Second Military Medical University, Shanghai, 200003, P. R. China
| | - Yiyun Cheng
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| |
Collapse
|
10
|
Maffucci T, Falasca M. Signalling Properties of Inositol Polyphosphates. Molecules 2020; 25:molecules25225281. [PMID: 33198256 PMCID: PMC7696153 DOI: 10.3390/molecules25225281] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/16/2022] Open
Abstract
Several studies have identified specific signalling functions for inositol polyphosphates (IPs) in different cell types and have led to the accumulation of new information regarding their cellular roles as well as new insights into their cellular production. These studies have revealed that interaction of IPs with several proteins is critical for stabilization of protein complexes and for modulation of enzymatic activity. This has not only revealed their importance in regulation of several cellular processes but it has also highlighted the possibility of new pharmacological interventions in multiple diseases, including cancer. In this review, we describe some of the intracellular roles of IPs and we discuss the pharmacological opportunities that modulation of IPs levels can provide.
Collapse
Affiliation(s)
- Tania Maffucci
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
- Correspondence: (T.M.); (M.F.); Tel.: +61-08-92669712 (M.F.)
| | - Marco Falasca
- School of Pharmacy and Biomedical Sciences, CHIRI, Curtin University, Perth 6102, Australia
- Correspondence: (T.M.); (M.F.); Tel.: +61-08-92669712 (M.F.)
| |
Collapse
|
11
|
Regulation of MicroRNA-155 and Its Related Genes Expression by Inositol Hexaphosphate in Colon Cancer Cells. Molecules 2019; 24:molecules24224153. [PMID: 31744065 PMCID: PMC6891702 DOI: 10.3390/molecules24224153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/06/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
Inositol hexaphosphate (IP6), a natural dietary component, has been found as an antitumor agent by stimulating apoptosis and inhibiting cancer cell proliferation, their migration, and metastasis in diverse cancers including colon cancer. However, molecular mechanisms of its action have not been well understood. In recent years, microRNAs (miRNAs) have been reported to play important roles in a broad range of biologic processes, such as cell growth, proliferation, apoptosis, or autophagy. These small noncoding molecules regulate post-transcriptional expression of targets genes via degradation of transcript or inhibition of protein synthesis. Aberrant expression and/or dysregulation of miRNAs have been characterized during tumor development and progression, thus, they are potential molecular targets for cancer prevention. The aim of this study was to investigate the effect of IP6 on the miRNAs expression profile in Caco-2 colon cancer cells. 84 miRNAs were analyzed in Caco-2 cells treated with 2.5 mM and 5 mM IP6 by the use of PCR (Polymerase Chain Reaction) array. The effect of 5 mM IP6 on selected potential miR-155 targets was determined by real-time (RT)-qPCR and ELISA (quantitative Polymerase Chain Reaction and Enzyme-Linked Immunosorbent Assay )method. The results indicated alteration in the specific 10 miRNA expression in human colon cancer cells following their treatment with 5 mM IP6. It down-regulated 8 miRNAs (miR-155, miR-210, miR-144, miR-194, miR-26b, miR-126, miR-302c, and miR-29a) and up-regulated 2 miRNAs (miR-223 and miR-196b). In silico analysis revealed that FOXO3a, HIF-1α, and ELK3 mRNAs are those of predicted targets of miR-155. IP6 at the concentration of 5 mM markedly induced FOXO3a and HIF-1a genes’ expression at both mRNA and protein level and decreased the amount of ELK3 mRNA as well as protein concentration in comparison to the control. In conclusion, the present study indicates that one of the mechanisms of antitumor potential of IP6 is down-regulation of the miR-155 expression in human colon cancer cells. Moreover, the expression of genes that are targeted by miRNA are also modulated by IP6.
Collapse
|
12
|
Inositol hexaphosphate plus inositol induced complete remission in stage IV melanoma: a case report. Melanoma Res 2019; 29:322-324. [PMID: 30615010 DOI: 10.1097/cmr.0000000000000577] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Inositol hexaphosphate (IP6) also called phytic acid is a polyphosphorylated carbohydrate naturally found in cereals, nuts, grains, and high-fiber-containing foods. It has been shown to inhibit the growth of many different tumor cell lines both in vitro and in vivo like colon, pancreas, liver, prostate, and even melanoma. Vitamin B inositol is a precursor of IP6 and another naturally occurring compound with anticancer properties. We present a case report of a patient with metastatic melanoma who declined traditional therapy and opted to try over the counter supplement IP6+inositol instead. To our surprise, the patient achieved a complete remission and remains in remission 3 years later. On the basis of this case and previous preclinical studies, we believe further research is indicated in exploring antiproliferative and potential immune stimulating effects of IP6+inositol in patients with metastatic melanoma.
Collapse
|
13
|
Anti-cancer activity of the cell membrane-permeable phytic acid prodrug. Bioorg Chem 2019; 92:103240. [PMID: 31525525 DOI: 10.1016/j.bioorg.2019.103240] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/11/2019] [Accepted: 08/30/2019] [Indexed: 02/02/2023]
Abstract
Phytic acid (IP6) is an ingredient in cereals and legumes, and limited amounts of this compound are considered to enter the cell and exert anti-cancer effects. These effects have been seen by studying cells treated with around 1-5 mM IP6. However, such a large amount of IP6 chelates metals and changes the pH in cell culture medium. To overcome this problem, we synthesized a prodrug of IP6 (Pro-IP6) and elucidated generation of IP6 from Pro-IP6 in cells. Cellular experiments using Pro-IP6 demonstrated selective anti-cancer effects including apoptosis and inhibition of Akt activation. Furthermore, an in vivo study using mice with adult T-cell leukemia also showed that Pro-IP6 reduced the size of the cancer. Taken together, Pro-IP6 is a useful biological tool and may lead to development of new anti-cancer drugs.
Collapse
|
14
|
Brehm MA, Windhorst S. New options of cancer treatment employing InsP 6. Biochem Pharmacol 2019; 163:206-214. [PMID: 30797871 DOI: 10.1016/j.bcp.2019.02.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/19/2019] [Indexed: 12/21/2022]
Abstract
Many mechanistic studies have been performed to analyze the cellular functions of the highly phosphorylated molecule inositol hexakisphosphate (InsP6) in health and disease. While the physiological intracellular functions are well described, the mechanism of potential pharmacological effects on cancer cell proliferation is still controversial. There are numerous studies demonstrating that a high InsP6 concentration (≥75 µM) inhibits growth of cancer cells in vitro and in vivo. Thus, there is no doubt that InsP6 exhibits anticancer activity but the mechanism underlying the cellular effects of extracellular InsP6 on cancer cells is far from being understood. In addition, studies on the inhibitory effect of InsP6 on cancer progression in animal models ignore aspects of its bioavailability. Here, we review and critically discuss the uptake mechanism and the intracellular involvement in signaling pathways of InsP6 in cancer cells. We take into account the controversial findings on InsP6 plasma concentration, which is a critical aspect of pharmacological accessibility of InsP6 for cancer treatment. Further, we discuss novel findings with respect to the effect of InsP6 on normal and immune cells as well as on platelet aggregate size. Our goal is to stimulate further mechanistic studies into novel directions considering previously disregarded aspects of InsP6. Only when we fully understand the mechanism underlying the anticancer activity of InsP6 novel and more efficient treatment options can be developed.
Collapse
Affiliation(s)
- Maria A Brehm
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany.
| |
Collapse
|
15
|
One stone with two birds: Phytic acid-capped platinum nanoparticles for targeted combination therapy of bone tumors. Biomaterials 2019; 194:130-138. [DOI: 10.1016/j.biomaterials.2018.12.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/12/2018] [Accepted: 12/21/2018] [Indexed: 12/17/2022]
|
16
|
Ren L, Hong ES, Mendoza A, Issaq S, Tran Hoang C, Lizardo M, LeBlanc A, Khanna C. Metabolomics uncovers a link between inositol metabolism and osteosarcoma metastasis. Oncotarget 2018; 8:38541-38553. [PMID: 28404949 PMCID: PMC5503552 DOI: 10.18632/oncotarget.15872] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 01/27/2017] [Indexed: 11/25/2022] Open
Abstract
Cancer development and progression are characterized by complex molecular events. The acquisition of these events is primarily believed to result from alterations in gene and protein expression/function. Recent studies have also suggested the role of metabolic alterations, or "metabolic reprogramming," that may similarly contribute to these events. Indeed, our previous investigations in osteosarcoma (OS) identified metabolic changes uniquely linked to metastasis. Based on those findings, here we sought to build a more detailed understanding of the specific alterations in metabolites or metabolic pathways that may be responsible for the observed metastasis-associated metabolic alterations, suggested by gene expression data. This was pursued using a combination of high-throughput liquid- and gas-chromatography-based mass spectrometry (LC/MS and GC/MS) for a global metabolic profiling/subtraction of four pairs of high/low metastatic OS cell lines. By comparing the identity and level of the metabolites between high/low metastatic cells, several metabolic pathways were identified to be differentially activated, such as arginine, glutathione, inositol and fatty acid metabolic pathways. To further interrogate these results, we investigated the effects of inositol pathway dysregulation, through the exposure of metastatic OS cells to IP6 (inositol hexaphosphate). Although IP6 exposures had modest to minimal effects on cell proliferation, we observed reduced cellular glycolysis, down-regulation of PI3K/Akt signaling and suppression of OS metastatic progression. Collectively these data supported further investigation of metabolic sensitivities as anti-metastatic strategies in a clinical setting as well as investigation of altered metabolomics associated with metastatic progression.
Collapse
Affiliation(s)
- Ling Ren
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Ellen S Hong
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Arnulfo Mendoza
- Molecular Oncology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Sameer Issaq
- Molecular Oncology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Christine Tran Hoang
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Michael Lizardo
- Molecular Oncology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Amy LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.,Dr. Khanna is currently with Ethos Veterinary Health, Woburn MA and Ethos Discovery, Washington DC, USA
| |
Collapse
|
17
|
Fu C, Tyagi R, Chin AC, Rojas T, Li RJ, Guha P, Bernstein IA, Rao F, Xu R, Cha JY, Xu J, Snowman AM, Semenza GL, Snyder SH. Inositol Polyphosphate Multikinase Inhibits Angiogenesis via Inositol Pentakisphosphate-Induced HIF-1α Degradation. Circ Res 2017; 122:457-472. [PMID: 29279301 DOI: 10.1161/circresaha.117.311983] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/18/2017] [Accepted: 12/22/2017] [Indexed: 12/17/2022]
Abstract
RATIONALE Inositol polyphosphate multikinase (IPMK) and its major product inositol pentakisphosphate (IP5) regulate a variety of cellular functions, but their role in vascular biology remains unexplored. OBJECTIVE We have investigated the role of IPMK in regulating angiogenesis. METHODS AND RESULTS Deletion of IPMK in fibroblasts induces angiogenesis in both in vitro and in vivo models. IPMK deletion elicits a substantial increase of VEGF (vascular endothelial growth factor), which mediates the regulation of angiogenesis by IPMK. The regulation of VEGF by IPMK requires its catalytic activity. IPMK is predominantly nuclear and regulates gene transcription. However, IPMK does not apparently serve as a transcription factor for VEGF. HIF (hypoxia-inducible factor)-1α is a major determinant of angiogenesis and induces VEGF transcription. IPMK deletion elicits a major enrichment of HIF-1α protein and thus VEGF. HIF-1α is constitutively ubiquitinated by pVHL (von Hippel-Lindau protein) followed by proteasomal degradation under normal conditions. However, HIF-1α is not recognized and ubiquitinated by pVHL in IPMK KO (knockout) cells. IP5 reinstates the interaction of HIF-1α and pVHL. HIF-1α prolyl hydroxylation, which is prerequisite for pVHL recognition, is interrupted in IPMK-deleted cells. IP5 promotes HIF-1α prolyl hydroxylation and thus pVHL-dependent degradation of HIF-1α. Deletion of IPMK in mouse brain increases HIF-1α/VEGF levels and vascularization. The increased VEGF in IPMK KO disrupts blood-brain barrier and enhances brain blood vessel permeability. CONCLUSIONS IPMK, via its product IP5, negatively regulates angiogenesis by inhibiting VEGF expression. IP5 acts by enhancing HIF-1α hydroxylation and thus pVHL-dependent degradation of HIF-1α.
Collapse
Affiliation(s)
- Chenglai Fu
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richa Tyagi
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alfred C Chin
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tomas Rojas
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ruo-Jing Li
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Prasun Guha
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Isaac A Bernstein
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Feng Rao
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Risheng Xu
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jiyoung Y Cha
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jing Xu
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Adele M Snowman
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Gregg L Semenza
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Solomon H Snyder
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
18
|
Inositol Hexaphosphate Inhibits Proliferation and Induces Apoptosis of Colon Cancer Cells by Suppressing the AKT/mTOR Signaling Pathway. Molecules 2017; 22:molecules22101657. [PMID: 28972559 PMCID: PMC6151581 DOI: 10.3390/molecules22101657] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/29/2017] [Accepted: 09/29/2017] [Indexed: 01/22/2023] Open
Abstract
AKT, a serine/threonine protein kinase and mammalian target of rapamycin (mTOR) plays a critical role in the proliferation and resistance to apoptosis that are essential to the development and progression of colon cancer. Therefore, AKT/mTOR signaling pathway has been recognized as an attractive target for anticancer therapy. Inositol hexaphosphate (InsP6), a natural occurring phytochemical, has been shown to have both preventive and therapeutic effects against various cancers, however, its exact molecular mechanisms of action are not fully understood. The aim of the in vitro study was to investigate the anticancer activity of InsP6 on colon cancer with the focus on inhibiting the AKT1 kinase and p70S6K1 as mTOR effector, in relation to proliferation and apoptosis of cells. The colon cancer Caco-2 cells were cultured using standard techniques and exposed to InsP6 at different concentrations (1 mM, 2.5 mM and 5 mM). Cellular proliferative activity was monitored by 5-bromo-2′-deoxyuridine (BrdU) incorporation into cellular DNA. Flow cytometric analysis was performed for cell cycle progression and apoptosis studies. Real-time RT-qPCR was used to validate mRNA levels of CDNK1A, CDNK1B, CASP3, CASP9, AKT1 and S6K1 genes. The concentration of p21 protein as well as the activities of caspase 3, AKT1 and p70S6K1 were determined by the ELISA method. The results revealed that IP6 inhibited proliferation and stimulated apoptosis of colon cancer cells. This effect was mediated by an increase in the expression of genes encoding p21, p27, caspase 3, caspase 9 as well a decrease in transcription of AKT1 and S6K1. InsP6 suppressed phosphorylation of AKT1 and p70S6K1, downstream effector of mTOR. Based on these studies it may be concluded that InsP6 can reduce proliferation and induce apoptosis through inhibition of the AKT/mTOR pathway and mTOR effector followed by modulation of the expression and activity of several key components of these pathways in colon cancer cells.
Collapse
|
19
|
Das F, Ghosh-Choudhury N, Kasinath BS, Choudhury GG. Tyrosines-740/751 of PDGFRβ contribute to the activation of Akt/Hif1α/TGFβ nexus to drive high glucose-induced glomerular mesangial cell hypertrophy. Cell Signal 2017; 42:44-53. [PMID: 28951244 DOI: 10.1016/j.cellsig.2017.09.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 09/22/2017] [Indexed: 01/24/2023]
Abstract
Glomerular mesangial cell hypertrophy contributes to the complications of diabetic nephropathy. The mechanism by which high glucose induces mesangial cell hypertrophy is poorly understood. Here we explored the role of the platelet-derived growth factor receptor-β (PDGFRβ) tyrosine kinase in driving the high glucose-induced mesangial cell hypertrophy. We show that high glucose stimulates the association of the PDGFRβ with PI 3 kinase leading to tyrosine phosphorylation of the latter. High glucose-induced Akt kinase activation was also dependent upon PDGFRβ and its tyrosine phosphorylation at 740/751 residues. Inhibition of PDGFRβ activity, its downregulation and expression of its phospho-deficient (Y740/751F) mutant inhibited mesangial cell hypertrophy by high glucose. Interestingly, expression of constitutively active Akt reversed this inhibition, indicating a role of Akt kinase downstream of PDGFRβ phosphorylation in this process. The transcription factor Hif1α is a target of Akt kinase. siRNAs against Hif1α inhibited the high glucose-induced mesangial cell hypertrophy. In contrast, increased expression of Hif1α induced hypertrophy similar to high glucose. We found that inhibition of PDGFRβ and expression of PDGFRβ Y740/751F mutant significantly inhibited the high glucose-induced expression of Hif1α. Importantly, expression of Hif1α countered the inhibition of mesangial cell hypertrophy induced by siPDGFRβ or PDGFRβ Y740/751F mutant. Finally, we show that high glucose-stimulated PDGFRβ tyrosine phosphorylation at 740/751 residues and the tyrosine kinase activity of the receptor regulate the transforming growth factor-β (TGFβ) expression by Hif1α. Thus we define the cell surface PDGFRβ as a major link between high glucose and its effectors Hif1α and TGFβ for induction of diabetic mesangial cell hypertrophy.
Collapse
Affiliation(s)
- Falguni Das
- Department of Medicine, UT Health at San Antonio, TX, United States
| | - Nandini Ghosh-Choudhury
- VA Biomedical Laboratory Research, South Texas Veterans Health Care System, San Antonio, TX, United States; Department of Pathology, UT Health at San Antonio, TX, United States
| | - Balakuntalam S Kasinath
- Department of Medicine, UT Health at San Antonio, TX, United States; VA Biomedical Laboratory Research, South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Goutam Ghosh Choudhury
- Department of Medicine, UT Health at San Antonio, TX, United States; VA Biomedical Laboratory Research, South Texas Veterans Health Care System, San Antonio, TX, United States; Geriatric Research, Education and Clinical Research, South Texas Veterans Health Care System, San Antonio, TX, United States.
| |
Collapse
|
20
|
Yu W, Liu C, Li X, Yang F, Cheng L, Liu C, Song Y. Inositol hexaphosphate suppresses colorectal cancer cell proliferation via the Akt/GSK-3β/β-catenin signaling cascade in a 1,2-dimethylhydrazine-induced rat model. Eur J Pharmacol 2017; 805:67-74. [DOI: 10.1016/j.ejphar.2017.03.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/02/2017] [Accepted: 03/10/2017] [Indexed: 01/31/2023]
|
21
|
Das F, Ghosh-Choudhury N, Venkatesan B, Kasinath BS, Ghosh Choudhury G. PDGF receptor-β uses Akt/mTORC1 signaling node to promote high glucose-induced renal proximal tubular cell collagen I (α2) expression. Am J Physiol Renal Physiol 2017; 313:F291-F307. [PMID: 28424212 DOI: 10.1152/ajprenal.00666.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 01/28/2023] Open
Abstract
Increased expression of PDGF receptor-β (PDGFRβ) has been shown in renal proximal tubules in mice with diabetes. The core molecular network used by high glucose to induce proximal tubular epithelial cell collagen I (α2) expression is poorly understood. We hypothesized that activation of PDGFRβ by high glucose increases collagen I (α2) production via the Akt/mTORC1 signaling pathway in proximal tubular epithelial cells. Using biochemical and molecular biological techniques, we investigated this hypothesis. We show that high glucose increases activating phosphorylation of the PDGFRβ, resulting in phosphorylation of phosphatidylinositol 3-kinase. A specific inhibitor, JNJ-10198409, and small interfering RNAs targeting PDGFRβ blocked this phosphorylation without having any effect on MEK/Erk1/2 activation. We also found that PDGFRβ regulates high glucose-induced Akt activation, its targets tuberin and PRAS40 phosphorylation, and finally, mTORC1 activation. Furthermore, inhibition of PDGFRβ suppressed high glucose-induced expression of collagen I (α2) in proximal tubular cells. Importantly, expression of constitutively active Akt or mTORC1 reversed these processes. As a mechanism, we found that JNJ and PDGFRβ knockdown inhibited high glucose-stimulated Hif1α expression. Furthermore, overexpression of Hif1α restored expression of collagen I (α2) that was inhibited by PDGFRβ knockdown in high glucose-stimulated cells. Finally, we show increased phosphorylation of PDGFRβ and its association with Akt/mTORC1 activation, Hif1α expression, and elevated collagen I (α2) levels in the renal cortex of mice with diabetes. Our results identify PDGFRβ as a driver in activating Akt/mTORC1 nexus for high glucose-mediated expression of collagen I (α2) in proximal tubular epithelial cells, which contributes to tubulointerstitial fibrosis in diabetic nephropathy.
Collapse
Affiliation(s)
- Falguni Das
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Nandini Ghosh-Choudhury
- VA Biomedical Laboratory Research, South Texas Veterans Health Care System, San Antonio, Texas.,Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Balachandar Venkatesan
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Balakuntalam S Kasinath
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas.,VA Biomedical Laboratory Research, South Texas Veterans Health Care System, San Antonio, Texas
| | - Goutam Ghosh Choudhury
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; .,VA Biomedical Laboratory Research, South Texas Veterans Health Care System, San Antonio, Texas.,Geriatric Research, Education and Clinical Research, South Texas Veterans Health Care System, San Antonio, Texas; and
| |
Collapse
|
22
|
Yin MX, Catimel B, Gregory M, Condron M, Kapp E, Holmes AB, Burgess AW. Synthesis of an inositol hexakisphosphate (IP6) affinity probe to study the interactome from a colon cancer cell line. Integr Biol (Camb) 2016; 8:309-18. [PMID: 26840369 DOI: 10.1039/c5ib00264h] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inositol hexakisphosphate (InsP6 or IP6) is an important signalling molecule in vesicular trafficking, neurotransmission, immune responses, regulation of protein kinases and phosphatases, activation of ion channels, antioxidant functions and anticancer activities. An IP6 probe was synthesised from myo-inositol via a derivatised analogue, which was immobilised through a terminal amino group onto Dynabeads. Systematic analysis of the IP6 interactome has been performed using the IP6 affinity probe using cytosolic extracts from the LIM1215 colonic carcinoma cell line. LC/MS/MS analysis identified 77 proteins or protein complexes that bind to IP6 specifically, including AP-2 complex proteins and β-arrestins as well as a number of novel potential IP6 interacting proteins. Bioinformatic enrichment analysis of the IP6 interactome reinforced the concept that IP6 regulates a number of biological processes including cell cycle and division, signal transduction, intracellular protein transport, vesicle-mediated transport and RNA splicing.
Collapse
Affiliation(s)
- Meng-Xin Yin
- School of Chemistry, Bio21 Institute, The University of Melbourne, 30 Flemington Road, Parkville, Victoria 3052, Australia
| | - Bruno Catimel
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Olivia Newton-John Cancer & Wellness Centre, Studley Road, Heidelberg, Victoria 3084, Australia
| | - Mark Gregory
- School of Chemistry, Bio21 Institute, The University of Melbourne, 30 Flemington Road, Parkville, Victoria 3052, Australia
| | - Melanie Condron
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia. and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Eugene Kapp
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia. and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Andrew B Holmes
- School of Chemistry, Bio21 Institute, The University of Melbourne, 30 Flemington Road, Parkville, Victoria 3052, Australia
| | - Antony W Burgess
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia. and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia and Department of Surgery, RMH, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
23
|
Oudart JB, Doué M, Vautrin A, Brassart B, Sellier C, Dupont-Deshorgue A, Monboisse JC, Maquart FX, Brassart-Pasco S, Ramont L. The anti-tumor NC1 domain of collagen XIX inhibits the FAK/ PI3K/Akt/mTOR signaling pathway through αvβ3 integrin interaction. Oncotarget 2016; 7:1516-28. [PMID: 26621838 PMCID: PMC4811477 DOI: 10.18632/oncotarget.6399] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 11/14/2015] [Indexed: 11/25/2022] Open
Abstract
Type XIX collagen is a minor collagen associated with basement membranes. It was isolated for the first time in a human cDNA library from rhabdomyosarcoma and belongs to the FACITs family (Fibril Associated Collagens with Interrupted Triple Helices). Previously, we demonstrated that the NC1 domain of collagen XIX (NC1(XIX)) exerts anti-tumor properties on melanoma cells by inhibiting their migration and invasion. In the present work, we identified for the first time the integrin αvβ3 as a receptor of NC1(XIX). Moreover, we demonstrated that NC1(XIX) inhibits the FAK/PI3K/Akt/mTOR pathway, by decreasing the phosphorylation and activity of the major proteins involved in this pathway. On the other hand, NC1(XIX) induced an increase of GSK3β activity by decreasing its degree of phosphorylation. Treatments targeting this central signaling pathway in the development of melanoma are promising and new molecules should be developed. NC1(XIX) seems to have the potential for the design of new anti-cancer drugs.
Collapse
Affiliation(s)
- Jean-Baptiste Oudart
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,CHU de Reims, Laboratoire Central de Biochimie, Reims, France
| | - Manon Doué
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Alexia Vautrin
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Bertrand Brassart
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Christèle Sellier
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Aurelie Dupont-Deshorgue
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Jean-Claude Monboisse
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,CHU de Reims, Laboratoire Central de Biochimie, Reims, France
| | - François-Xavier Maquart
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,CHU de Reims, Laboratoire Central de Biochimie, Reims, France
| | - Sylvie Brassart-Pasco
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Laurent Ramont
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,CHU de Reims, Laboratoire Central de Biochimie, Reims, France
| |
Collapse
|
24
|
Thamilselvan V, Menon M, Thamilselvan S. Combination of carmustine and selenite effectively inhibits tumor growth by targeting androgen receptor, androgen receptor-variants, and Akt in preclinical models: New hope for patients with castration resistant prostate cancer. Int J Cancer 2016; 139:1632-47. [PMID: 27198552 DOI: 10.1002/ijc.30189] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 04/28/2016] [Accepted: 05/03/2016] [Indexed: 01/12/2023]
Abstract
Despite established androgen receptor (AR) antagonists, AR/AR-variants signaling remain a major obstacle for the successful treatment of castration resistant prostate cancer (CRPC). In addition, CRPC cells adapt to survive via AR-independent pathways to escape next generation therapies. Therefore, there is an urgent need for drugs that can target these signaling pathways in CRPC. In this study, we sought to determine whether carmustine and selenite in combination could induce apoptosis and inhibit growth of CRPC in-vitro and in-vivo. CRPC (22Rv1, VCaP, and PC-3) cell lines in culture and xenograft mouse were used. Combination of carmustine and selenite treatment significantly increased reactive oxygen species, apoptosis and growth inhibition in CRPC cells with down regulation of anti-apoptotic (Bcl-2 and Mcl-1) and proliferative proteins (c-Myc and cyclin-D1). This effect was associated with complete reduction of AR/AR-variants, AR-V7, PSA and significant induction of p27Kip1. Combination treatment substantially abolished phospho-Akt, phospho-GSK-3β, and anchorage-independent growth in AR-positive and AR-negative cells. Consistent with in-vitro results, combination treatment effectively induced apoptosis and completely inhibited xenograft tumor growth and markedly reduced AR/AR-variants, AR-V7, PSA, and Bcl-2 in xenograft tumors without causing genotoxicity in host mice. Individual agent treatment showed only partial effect. The combination treatment showed a significant synergistic effect. The present study is the first to demonstrate that the combination of carmustine and selenite treatment completely suppressed CRPC tumor growth by reducing AR/AR-variants and Akt signaling. Our findings suggest that the combination of carmustine and selenite could constitute a promising next-generation therapy for successful treatment of patients with CRPC.
Collapse
Affiliation(s)
| | - Mani Menon
- Vattikuti Urology Institute, Henry Ford Health System, Detroit, MI
| | | |
Collapse
|
25
|
Bizzarri M, Dinicola S, Bevilacqua A, Cucina A. Broad Spectrum Anticancer Activity of Myo-Inositol and Inositol Hexakisphosphate. Int J Endocrinol 2016; 2016:5616807. [PMID: 27795708 PMCID: PMC5067332 DOI: 10.1155/2016/5616807] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/06/2016] [Indexed: 02/06/2023] Open
Abstract
Inositols (myo-inositol and inositol hexakisphosphate) exert a wide range of critical activities in both physiological and pathological settings. Deregulated inositol metabolism has been recorded in a number of diseases, including cancer, where inositol modulates different critical pathways. Inositols inhibit pRB phosphorylation, fostering the pRB/E2F complexes formation and blocking progression along the cell cycle. Inositols reduce PI3K levels, thus counteracting the activation of the PKC/RAS/ERK pathway downstream of PI3K activation. Upstream of that pathway, inositols disrupt the ligand interaction between FGF and its receptor as well as with the EGF-transduction processes involving IGF-II receptor and AP-1 complexes. Additionally, Akt activation is severely impaired upon inositol addition. Downregulation of both Akt and ERK leads consequently to NF-kB inhibition and reduced expression of inflammatory markers (COX-2 and PGE2). Remarkably, inositol-induced downregulation of presenilin-1 interferes with the epithelial-mesenchymal transition and reduces Wnt-activation, β-catenin translocation, Notch-1, N-cadherin, and SNAI1 release. Inositols interfere also with the cytoskeleton by upregulating Focal Adhesion Kinase and E-cadherin and decreasing Fascin and Cofilin, two main components of pseudopodia, leading hence to invasiveness impairment. This effect is reinforced by the inositol-induced inhibition on metalloproteinases and ROCK1/2 release. Overall, these effects enable inositols to remodel the cytoskeleton architecture.
Collapse
Affiliation(s)
- Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Lab, Sapienza University of Rome, Rome, Italy
- *Mariano Bizzarri:
| | - Simona Dinicola
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy
| | - Arturo Bevilacqua
- Department of Psychology, Section of Neuroscience, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
| | - Alessandra Cucina
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy
- Azienda Policlinico Umberto I, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
26
|
Sheng T, Shen RL, Shao H, Ma TH. No association between fiber intake and prostate cancer risk: a meta-analysis of epidemiological studies. World J Surg Oncol 2015; 13:264. [PMID: 26315558 PMCID: PMC4552444 DOI: 10.1186/s12957-015-0681-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/17/2015] [Indexed: 11/17/2022] Open
Abstract
Background The findings of epidemiologic studies on the association between fiber intake and prostate cancer risk remain conflicting. We aimed to examine this association by conducting a meta-analysis of epidemiological studies. Methods Relevant studies were identified by PubMed (1966 to March 2015) and Embase (1974 to March 2015) database search through March 2015. We included epidemiological studies that reported relative risks (RRs) or odds ratios (ORs) with 95 % confidence intervals (CIs) for the association between dietary fiber intake and prostate cancer risk. Random effects models were used to calculate the summary risk estimates. Results For the highest compared with the lowest dietary fiber intake, a significantly decreased risk with prostate cancer was observed in case-control studies (OR = 0.82; 95 % CI, 0.68–0.96), but not in cohort studies (RR = 0.94; 95 % CI, 0.77–1.11). The combined risk estimate of all studies was 0.89 (95 % CI, 0.77, 1.01). A significant heterogeneity was observed across studies (p = 0.005). There was no evidence of significant publication bias based on Begg’s funnel plot (p = 0.753) or Egger’s test (p = 0.946). Conclusions This meta-analysis suggests the absence of evidence for association between dietary fiber intake and prostate cancer risk.
Collapse
Affiliation(s)
- Tao Sheng
- Department of Urology, Jiaxing Affilated Hospital of Zhejiang Chinese Medical University, Jiaxing, Zhejiang Province, China.
| | - Rui-lin Shen
- Department of Urology, Jiaxing Affilated Hospital of Zhejiang Chinese Medical University, Jiaxing, Zhejiang Province, China.
| | - Huan Shao
- Department of Urology, Jiaxing Affilated Hospital of Zhejiang Chinese Medical University, Jiaxing, Zhejiang Province, China.
| | - Tian-hong Ma
- Department of Pharmacy, Jiaxing Affilated Hospital of Zhejiang Chinese Medical University, Zhongshan East Road 1501, Jiaxing, Zhejiang Province, 314001, China.
| |
Collapse
|
27
|
Sahay S, Tiwari P, Gupta KP. Onset of the lymphocytic infiltration and hyperplasia preceding the proliferation in F1 mouse lungs from the N-ethyl-N-nitrosourea exposed mothers: Prevention during the lactation period by inositol hexaphosphate. Toxicol Rep 2015; 2:590-599. [PMID: 28962394 PMCID: PMC5598482 DOI: 10.1016/j.toxrep.2015.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/23/2015] [Accepted: 03/23/2015] [Indexed: 12/04/2022] Open
Abstract
Maternal exposure to a carcinogen is associated with increased risk of different cancers in the offspring. The foetus is highly sensitive to carcinogens and this contributes to the foetal basis of the onset of disease. The better understanding of the molecular mechanisms involved in the early stage of lung tumourigenesis in the offspring is needed for the newer preventive strategies. We evaluated the effects of N-ethyl-N-nitrosourea (ENU) given on the 17th day of gestation and antitumour agent inositol hexaphosphate (IP6) to the mothers at the early stage of lung tumourigenesis in F1 mice. There was no treatment related effects on the litter size or body weight of the F1 mice at the PND12 or 24. Analysis of PCNA, NF-κB (p50), IL-6, COX-2, pSTAT3, STAT3, caspase-3, caspase-9, PARP, Akt signalling and downstream cyclin D1 along with miR-155, suggested the modulation of proliferation, inflammation and apoptosis at PND12 and 24. IP6 administration to the predisposed mothers prevented the proliferation, inflammation and enhanced apoptosis in F1 lung as showed by a reduction in PCNA, NF-κB (p50), IL-6, COX-2, pSTAT3, STAT3, miR-155 and increase in caspases, cleavage of poly (ADP-ribose) polymerase. IP6 administration also inhibited the activation of Akt and cyclin D1. Our study shows that tumourigenic changes take place in the lungs of the F1 generation from the carcinogen predisposed mothers even before the onset of tumours and the simultaneous intake of chemopreventive agent during the gestation or lactation period could prevent the lymphocytic infiltration and hyperplasia preceding the tumourigenesis.
Collapse
Affiliation(s)
- Satya Sahay
- Environmental Carcinogenesis Laboratory, CSIR-Indian Institute of Toxicology Research, Mahatma Gandhi Marg, Lucknow 226001, India
| | - Prakash Tiwari
- Environmental Carcinogenesis Laboratory, CSIR-Indian Institute of Toxicology Research, Mahatma Gandhi Marg, Lucknow 226001, India
| | - Krishna P Gupta
- Environmental Carcinogenesis Laboratory, CSIR-Indian Institute of Toxicology Research, Mahatma Gandhi Marg, Lucknow 226001, India
| |
Collapse
|
28
|
Liu G, Song Y, Cui L, Wen Z, Lu X. Inositol hexaphosphate suppresses growth and induces apoptosis in HT-29 colorectal cancer cells in culture: PI3K/Akt pathway as a potential target. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:1402-1410. [PMID: 25973024 PMCID: PMC4396237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 01/23/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Inositol hexaphosphate (IP6) is a polyphosphorylated carbohydrate that is present in high amounts in almost all plants and mammalian cells. IP6 induces apoptosis in multiple types of cancer cells, including prostate cancer, breast cancer, skin tumor, liver cancer and colorectal cancer. However, little is known regarding the molecular mechanisms of its anticancer effects. Therefore, this study was conducted to investigate the activity of IP6 against human colorectal cancer cells (HT-29) and to determine whether the IP6 regulates apoptosis in HT-29 cells by inhibiting the PI3K/Akt signaling pathway. METHOD A human colorectal cancer cell line (HT-29) was used for the study. HT-29 cells were treated with 0, 50, 100, 200, and 400 μg/mL of IP6. The MTT colorimetric assay was used to observe the proliferation of HT-29 in vitro, and flow cytometry (FCM) was used to analyze the apoptosis of the HT-29 cells. The relative mRNA expression was determined by real-time PCR, and relative protein levels were analyzed by Western blot analysis. RESULT The results of MTT showed that HT-29 cells underwent inhibition of proliferation after exposure to IP6 (100-400 μg/mL) for 12 and 48 h, and this inhibition clearly relied on time and dosage. IP6 induced apoptosis in HT-29 cells in a dose-dependent manner. The mRNA and protein expression of PI3K and Akt decreased in the groups treated with IP6, and IP6 inhibited the phosphorylation of Akt (pAkt), whereas increased the expression of its downstream effector, caspase-9. CONCLUSION Our results suggested that by targeting PI3K/Akt pathway, IP6 suppresses cell survival and proliferation, but induces death in HT-29 cells.
Collapse
Affiliation(s)
- Guiyuan Liu
- Department of Public Health, Qingdao University Medical CollegeQingdao 266021, Shangdong, China
| | - Yang Song
- Department of Public Health, Qingdao University Medical CollegeQingdao 266021, Shangdong, China
| | - Lianhua Cui
- Department of Public Health, Qingdao University Medical CollegeQingdao 266021, Shangdong, China
| | - Zhaoxia Wen
- School of Nursing, Qingdao University Medical CollegeQingdao 266021, Shangdong, China
| | - Xiaoqing Lu
- Department of Public Health, Qingdao University Medical CollegeQingdao 266021, Shangdong, China
| |
Collapse
|
29
|
Ji H, Li Y, Jiang F, Wang X, Zhang J, Shen J, Yang X. Inhibition of transforming growth factor beta/SMAD signal by MiR-155 is involved in arsenic trioxide-induced anti-angiogenesis in prostate cancer. Cancer Sci 2014; 105:1541-9. [PMID: 25283513 PMCID: PMC4317958 DOI: 10.1111/cas.12548] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 09/24/2014] [Accepted: 09/30/2014] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer is the most common cause of cancer-related deaths in men. Current practices for treatment of prostate cancer are less than satisfactory because of metastasis and recurrence, which are primarily attributed to angiogenesis. Hence, anti-angiogenesis treatment is becoming a promising new approach for prostate cancer therapy. In addition to treating acute promyelocytic leukemia, arsenic trioxide (As2O3) suppresses other solid tumors, including prostate cancer. However, the effects of As2O3 on angiogenesis in prostate cancer cells, and the underlying molecular mechanisms remain unclear. In the present study, As2O3 attenuated angiogenic ability through microRNA-155 (miR-155)-mediated inhibition of transforming growth factor beta (TGF-β)/SMAD signal pathway in human prostate cancer PC-3 and LNCaP cells in vitro and in vivo. Briefly, As2O3 inhibited the activations/expressions of both TGFβ-induced and endogenous SMAD2/3. Furthermore, As2O3 improved the expression of miR-155 via DNA-demethylation. MiR-155, which targeted the SMAD2-3′UTR, decreased the expression and function of SMAD2. Knockdown of miR-155 abolished the As2O3-induced inhibitions of the TGF-β/SMAD2 signaling, the vascular endothelial growth factor secretion and angiogenesis. Through understanding a novel mechanism whereby As2O3 inhibits angiogenic potential of prostate cancer cells, our study would help in the development of As2O3 as a potential chemopreventive agent when used alone or in combination with other current anticancer drugs.
Collapse
Affiliation(s)
- Hui Ji
- Affiliated Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
30
|
Raditic DM, Bartges JW. Evidence-based Integrative Medicine in Clinical Veterinary Oncology. Vet Clin North Am Small Anim Pract 2014; 44:831-53. [DOI: 10.1016/j.cvsm.2014.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
31
|
Shang D, Li C, Yao Q, Yang H, Xu Y, Han J, Li J, Su F, Zhang Y, Zhang C, Li D, Li X. Prioritizing candidate disease metabolites based on global functional relationships between metabolites in the context of metabolic pathways. PLoS One 2014; 9:e104934. [PMID: 25153931 PMCID: PMC4143229 DOI: 10.1371/journal.pone.0104934] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 07/14/2014] [Indexed: 11/18/2022] Open
Abstract
Identification of key metabolites for complex diseases is a challenging task in today's medicine and biology. A special disease is usually caused by the alteration of a series of functional related metabolites having a global influence on the metabolic network. Moreover, the metabolites in the same metabolic pathway are often associated with the same or similar disease. Based on these functional relationships between metabolites in the context of metabolic pathways, we here presented a pathway-based random walk method called PROFANCY for prioritization of candidate disease metabolites. Our strategy not only takes advantage of the global functional relationships between metabolites but also sufficiently exploits the functionally modular nature of metabolic networks. Our approach proved successful in prioritizing known metabolites for 71 diseases with an AUC value of 0.895. We also assessed the performance of PROFANCY on 16 disease classes and found that 4 classes achieved an AUC value over 0.95. To investigate the robustness of the PROFANCY, we repeated all the analyses in two metabolic networks and obtained similar results. Then we applied our approach to Alzheimer's disease (AD) and found that a top ranked candidate was potentially related to AD but had not been reported previously. Furthermore, our method was applicable to prioritize the metabolites from metabolomic profiles of prostate cancer. The PROFANCY could identify prostate cancer related-metabolites that are supported by literatures but not considered to be significantly differential by traditional differential analysis. We also developed a freely accessible web-based and R-based tool at http://bioinfo.hrbmu.edu.cn/PROFANCY.
Collapse
Affiliation(s)
- Desi Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Chunquan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, P. R. China
| | - Qianlan Yao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Haixiu Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Yanjun Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Jing Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Fei Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Dongguo Li
- School of Biomedical Engineering, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Beijing, P.R. China
- * E-mail: (DL); (XL)
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
- * E-mail: (DL); (XL)
| |
Collapse
|
32
|
Varna M, Bertheau P, Legrès L. Tumor Microenvironment in Human Tumor Xenografted Mouse Models. ACTA ACUST UNITED AC 2014. [DOI: 10.6000/1927-7229.2014.03.03.6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
33
|
Wang YL, Zhu BJ, Qi ZZ, Wang HJ, Zhou XD. Akt1 enhances CA916798 expression through mTOR pathway. PLoS One 2013; 8:e62327. [PMID: 23667466 PMCID: PMC3648559 DOI: 10.1371/journal.pone.0062327] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/20/2013] [Indexed: 01/07/2023] Open
Abstract
Multi-drug resistance leads to the failure of chemotherapy for cancers. Our previous study showed that overexpression of CA916798 led to multi-drug resistance. However, the underlying mechanisms remain unknown. In the current study, we observed that the levels of phosphorylated AKT, phosphorylated mTOR and CA916798 all increased in the drug resistant human adenocarcinoma samples and paralleled with the change of drug resistance. The results of immunofluorescence and Co-IP indicated that the positive correlation of CA916798 expression with AKT1 activation might be associated with drug resistance of lung adenocarcinoma. Furthermore, AKT1 stimulated CA916798 expression through mTOR pathway in both A549 and A549/CDDP cell lines, which was also observed in the xenografted tumor in nude mice. The results showed that CA916798 located in the downstream of PI3K/AKT/mTOR pathway. Inhibition of PI3K by LY294002 could efficiently reduce CA916798 expression and tumor size in vivo as well. Additionally, LY294002 combined with rapamycin inhibited CA916798 expression and tumor size stronger than LY294002 alone. Our findings may also provide a new explanation for synergistic anti-tumor effects of PI3K and mTORC1 inhibitors.
Collapse
Affiliation(s)
- Yu-Liang Wang
- Department of Respiratory Medicine, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Bing-Jing Zhu
- Department of Respiratory Medicine, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Zhan-Zhong Qi
- Department of Respiratory Medicine, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Hai-Jing Wang
- Department of Respiratory Medicine, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Xiang-Dong Zhou
- Department of Respiratory Medicine, Southwest Hospital, The Third Military Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
34
|
Raina K, Ravichandran K, Rajamanickam S, Huber KM, Serkova NJ, Agarwal R. Inositol hexaphosphate inhibits tumor growth, vascularity, and metabolism in TRAMP mice: a multiparametric magnetic resonance study. Cancer Prev Res (Phila) 2012; 6:40-50. [PMID: 23213071 DOI: 10.1158/1940-6207.capr-12-0387] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Herein, employing anatomical and dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI), we evaluated noninvasively, the in vivo, chemopreventive efficacy of inositol hexaphosphate (IP6), a major constituent of high-fiber diets, against prostate tumor growth and progression in transgenic adenocarcinoma of the mouse prostate (TRAMP) model. Male TRAMP mice, beginning at 4 weeks of age, were fed with 1%, 2%, or 4% (w/v) IP6 in drinking water or only drinking water till 28 weeks of age and monitored using MRI over the course of study. Longitudinal assessment of prostate volumes by conventional MRI and tumor vascularity by gadolinium-based DCE-MRI showed a profound reduction in tumor size, partly due to antiangiogenic effects by IP6 treatment. As potential mechanisms of IP6 efficacy, decrease in the expression of glucose transporter GLUT-4 protein together with an increase in levels of phospho-AMP-activated kinase (AMPK(Th172)) were observed in prostate tissues of mice from IP6 fed-groups, suggesting that IP6 is interfering with the metabolic events occurring in TRAMP prostate. Investigative metabolomics study using quantitative high-resolution (1)H-NMR on prostate tissue extracts showed that IP6 significantly decreased glucose metabolism and membrane phospholipid synthesis, in addition to causing an increase in myoinositol levels in the prostate. Together, these findings show that oral IP6 supplement blocks growth and angiogenesis of prostate cancer in the TRAMP model in conjunction with metabolic events involved in tumor sustenance. This results in energy deprivation within the tumor, suggesting a practical and translational potential of IP6 treatment in suppressing growth and progression of prostate cancer in humans.
Collapse
Affiliation(s)
- Komal Raina
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | | | | | | |
Collapse
|
35
|
Tyagi A, Gu M, Takahata T, Frederick B, Agarwal C, Siriwardana S, Agarwal R, Sclafani RA. Resveratrol selectively induces DNA Damage, independent of Smad4 expression, in its efficacy against human head and neck squamous cell carcinoma. Clin Cancer Res 2011; 17:5402-11. [PMID: 21705453 DOI: 10.1158/1078-0432.ccr-11-1072] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Alterations in Smad4 signaling and its loss cause genomic instability and head and neck squamous cell carcinoma (HNSCC), suggesting that agents that target both Smad4-dependent and -independent pathways could control HNSCC. EXPERIMENTAL DESIGN Resveratrol efficacy was evaluated against the HNSCC cells FaDu, Cal27, Det562, and Cal27-Smad4 for viability, DNA damage, cell-cycle progression, and apoptosis, as well as γ-H2AX expression, and focus formation (γ-H2AX and Brca1). Resveratrol efficacy was also examined in nude mice for FaDu xenograft growth. Xenografts were analyzed for γ-H2AX and cleaved caspase-3. RESULTS Resveratrol (5-50 μmol/L) suppressed viability and induced DNA damage in FaDu and Cal27 cells but not in normal human epidermal keratinocytes and human foreskin fibroblasts, showing its selectivity toward HNSCC cells; however, Det562 cells were resistant to resveratrol even at 100 μmol/L. Cal27 cells stably transfected with Smad4 showed similar resveratrol effects as parental Cal27, indicating that a lack of resveratrol effect in Det562 cells was independent of Smad4 status in these cells. Furthermore, resveratrol caused S-phase arrest and apoptotic death of FaDu and Cal27 cells together with induction of Brca1 and γ-H2AX foci. Resveratrol (50 mg/kg body weight) treatment also inhibited FaDu tumor growth in nude mice, and γ-H2AX and cleaved caspase-3 were strongly increased in xenografts from resveratrol-treated mice compared with controls. CONCLUSION Our findings for the first time showed antiproliferative, DNA damaging, and apoptotic effects of resveratrol in HNSCC cells independent of Smad4 status, both in vitro and in vivo, suggesting that more studies are needed to establish its potential usefulness against HNSCC.
Collapse
Affiliation(s)
- Alpna Tyagi
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Stenman K, Stattin P, Stenlund H, Riklund K, Gröbner G, Bergh A. H HRMAS NMR Derived Bio-markers Related to Tumor Grade, Tumor Cell Fraction, and Cell Proliferation in Prostate Tissue Samples. Biomark Insights 2011; 6:39-47. [PMID: 21499438 PMCID: PMC3076017 DOI: 10.4137/bmi.s6794] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
A high-resolution magic angle spinning NMR spectroscopic approach is presented for evaluating the occurrence, amount and aggressiveness of cancer in human prostate tissue samples. Using this technique, key metabolites in malignant and non-malignant samples (n = 149) were identified, and patterns of their relative abundance were analyzed by multivariate statistical methods. Ratios of various metabolites – including (glycerophophorylcholine + phosphorylcholine)/creatine, myo-inositol/scyllo-inositol, scyllo-inositol/creatine, choline/creatine, and citrate/creatine – correlated with: i) for non-malignant tissue samples, the distance to the nearest tumor and its Gleason score and; ii) the fraction of tumor cells present in the sample; and iii) tumor cell proliferation (Ki67 labelling index). This NMR-based approach allows the extraction of information that could be useful for developing novel diagnostic methods for prostate cancer.
Collapse
|
37
|
Ravichandran K, Velmurugan B, Gu M, Singh RP, Agarwal R. Inhibitory effect of silibinin against azoxymethane-induced colon tumorigenesis in A/J mice. Clin Cancer Res 2010; 16:4595-606. [PMID: 20823143 DOI: 10.1158/1078-0432.ccr-10-1213] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Colorectal cancer is the second leading cause of cancer-associated deaths, which suggests that more effort is needed to prevent/control this disease. Herein, for the first time, we investigate in vivo the efficacy of silibinin against azoxymethane-induced colon tumorigenesis in A/J mice. EXPERIMENTAL DESIGN Five-week-old male mice were gavaged with vehicle or silibinin (250 and 750 mg/kg) for 25 weeks starting 2 weeks before initiation with azoxymethane (pretreatment regime) or for 16 weeks starting 2 weeks after the last azoxymethane injection (posttreatment regime). The mice were then sacrificed, and colon tissues were examined for tumor multiplicity and size, and molecular markers for proliferation, apoptosis, inflammation, and angiogenesis. RESULTS Silibinin feeding showed a dose-dependent decrease in azoxymethane-induced colon tumorigenesis with stronger efficacy in pretreatment versus posttreatment regimen. Mechanistic studies in tissue samples showed that silibinin inhibits cell proliferation as evident by a decrease (P < 0.001) in proliferating cell nuclear antigen and cyclin D1, and increased Cip1/p21 levels. Silibinin also decreased (P < 0.001) the levels of inducible nitric oxide synthase, cyclooxygenase-2, and vascular endothelial growth factor, suggesting its anti-inflammatory and antiangiogenic potential in this model. Further, silibinin increased cleaved caspase-3 and poly(ADP-ribose) polymerase levels, indicating its apoptotic effect. In other studies, colonic mucosa and tumors expressed high levels of β-catenin, insulin-like growth factor-1 receptorβ, phospho Glycogen synthase kinase-3β, and phospho protein kinase B/pAkt proteins in azoxymethane-treated mice, which were strongly lowered (P < 0.001) by silibinin treatment. Moreover, azoxymethane reduced insulin-like growth factor binding protein-3 protein level, which was enhanced by silibinin. CONCLUSIONS Silibinin targets β-catenin and IGF-1Rβ pathways for its chemopreventive efficacy against azoxymethane-induced colon carcinogenesis in A/J mice. Overall, these results support the translational potential of silibinin in colorectal cancer chemoprevention.
Collapse
Affiliation(s)
- Kameswaran Ravichandran
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, Aurora, Colorado 80045, USA
| | | | | | | | | |
Collapse
|