1
|
Fatima I, Ahmad R, Barman S, Gowrikumar S, Pravoverov K, Primeaux M, Fisher KW, Singh AB, Dhawan P. Albendazole inhibits colon cancer progression and therapy resistance by targeting ubiquitin ligase RNF20. Br J Cancer 2024; 130:1046-1058. [PMID: 38278978 PMCID: PMC10951408 DOI: 10.1038/s41416-023-02570-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND The repurposing of FDA-approved drugs for anti-cancer therapies is appealing due to their established safety profiles and pharmacokinetic properties and can be quickly moved into clinical trials. Cancer progression and resistance to conventional chemotherapy remain the key hurdles in improving the clinical management of colon cancer patients and associated mortality. METHODS High-throughput screening (HTS) was performed using an annotated library of 1,600 FDA-approved drugs to identify drugs with strong anti-CRC properties. The candidate drug exhibiting most promising inhibitory effects in in-vitro studies was tested for its efficacy using in-vivo models of CRC progression and chemoresistance and patient derived organoids (PTDOs). RESULTS Albendazole, an anti-helminth drug, demonstrated the strongest inhibitory effects on the tumorigenic potentials of CRC cells, xenograft tumor growth and organoids from mice. Also, albendazole sensitized the chemoresistant CRC cells to 5-fluorouracil (5-FU) and oxaliplatin suggesting potential to treat chemoresistant CRC. Mechanistically, Albendazole treatment modulated the expression of RNF20, to promote apoptosis in CRC cells by delaying the G2/M phase and suppressing anti-apoptotic-Bcl2 family transcription. CONCLUSIONS Albendazole, an FDA approved drug, carries strong therapeutic potential to treat colon cancers which are aggressive and potentially resistant to conventional chemotherapeutic agents. Our findings also lay the groundwork for further clinical testing.
Collapse
Affiliation(s)
- Iram Fatima
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Susmita Barman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saiprasad Gowrikumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kristina Pravoverov
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mark Primeaux
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kurt W Fisher
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
2
|
Tincu (Iurciuc) CE, Andrițoiu CV, Popa M, Ochiuz L. Recent Advancements and Strategies for Overcoming the Blood-Brain Barrier Using Albumin-Based Drug Delivery Systems to Treat Brain Cancer, with a Focus on Glioblastoma. Polymers (Basel) 2023; 15:3969. [PMID: 37836018 PMCID: PMC10575401 DOI: 10.3390/polym15193969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive malignant tumor, and the most prevalent primary malignant tumor affecting the brain and central nervous system. Recent research indicates that the genetic profile of GBM makes it resistant to drugs and radiation. However, the main obstacle in treating GBM is transporting drugs through the blood-brain barrier (BBB). Albumin is a versatile biomaterial for the synthesis of nanoparticles. The efficiency of albumin-based delivery systems is determined by their ability to improve tumor targeting and accumulation. In this review, we will discuss the prevalence of human glioblastoma and the currently adopted treatment, as well as the structure and some essential functions of the BBB, to transport drugs through this barrier. We will also mention some aspects related to the blood-tumor brain barrier (BTBB) that lead to poor treatment efficacy. The properties and structure of serum albumin were highlighted, such as its role in targeting brain tumors, as well as the progress made until now regarding the techniques for obtaining albumin nanoparticles and their functionalization, in order to overcome the BBB and treat cancer, especially human glioblastoma. The albumin drug delivery nanosystems mentioned in this paper have improved properties and can overcome the BBB to target brain tumors.
Collapse
Affiliation(s)
- Camelia-Elena Tincu (Iurciuc)
- Department of Natural and Synthetic Polymers, “Cristofor Simionescu” Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 73, Prof. Dimitrie Mangeron Street, 700050 Iasi, Romania;
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16, University Street, 700115 Iasi, Romania;
| | - Călin Vasile Andrițoiu
- Apitherapy Medical Center, Balanesti, Nr. 336-337, 217036 Gorj, Romania;
- Specialization of Nutrition and Dietetics, Faculty of Pharmacy, Vasile Goldis Western University of Arad, Liviu Rebreanu Street, 86, 310045 Arad, Romania
| | - Marcel Popa
- Department of Natural and Synthetic Polymers, “Cristofor Simionescu” Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 73, Prof. Dimitrie Mangeron Street, 700050 Iasi, Romania;
- Faculty of Dental Medicine, “Apollonia” University of Iasi, 11, Pacurari Street, 700511 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania
| | - Lăcrămioara Ochiuz
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16, University Street, 700115 Iasi, Romania;
| |
Collapse
|
3
|
Zhang H, Luo P, Huang X. Engineered nanomaterials enhance drug delivery strategies for the treatment of osteosarcoma. Front Pharmacol 2023; 14:1269224. [PMID: 37670948 PMCID: PMC10475588 DOI: 10.3389/fphar.2023.1269224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in adolescents, and the clinical treatment of OS mainly includes surgery, radiotherapy, and chemotherapy. However, the side effects of chemotherapy drugs are an issue that clinicians cannot ignore. Nanomedicine and drug delivery technologies play an important role in modern medicine. The development of nanomedicine has ushered in a new turning point in tumor treatment. With the emergence and development of nanoparticles, nanoparticle energy surfaces can be designed with different targeting effects. Not only that, nanoparticles have unique advantages in drug delivery. Nanoparticle delivery drugs can not only reduce the toxic side effects of chemotherapy drugs, but due to the enhanced permeability retention (EPR) properties of tumor cells, nanoparticles can survive longer in the tumor microenvironment and continuously release carriers to tumor cells. Preclinical studies have confirmed that nanoparticles can effectively delay tumor growth and improve the survival rate of OS patients. In this manuscript, we present the role of nanoparticles with different functions in the treatment of OS and look forward to the future treatment of improved nanoparticles in OS.
Collapse
Affiliation(s)
- Haorui Zhang
- Department of Spine, Trauma Surgery, The First People’s Hospital of Guangyuan, Guangyuan, China
| | - Ping Luo
- Science and Technology Education Section, The First People’s Hospital of Guangyuan, Guangyuan, China
| | - Xiaojun Huang
- Department of Spine, Trauma Surgery, The First People’s Hospital of Guangyuan, Guangyuan, China
| |
Collapse
|
4
|
Wu K, Peng X, Chen M, Li Y, Tang G, Peng J, Peng Y, Cao X. Recent progress of research on anti‐tumor agents using benzimidazole as the structure unit. Chem Biol Drug Des 2022; 99:736-757. [DOI: 10.1111/cbdd.14022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/10/2022] [Accepted: 01/15/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Kaiyue Wu
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Xiaoyu Peng
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Miaojia Chen
- Department of Pharmacy the first People's Hospital Pingjiang Yueyang Hunan China
| | - Yang Li
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Junmei Peng
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Yuanyuan Peng
- School of Electrical and Automation Engineering East China Jiaotong University Nanchang 330000 China
| | - Xuan Cao
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| |
Collapse
|
5
|
El-Dardiry MA, Abdel-Aal AA, Abdeltawab MSA, El-Sherbini M, Hassan MA, Abdel-Aal AA, Badawi M, Anis SE, Khaled BEA, Al-Antably AS. Effect of mast cell stabilization on angiogenesis in primary and secondary experimental Trichinella spiralis infection. Parasit Vectors 2021; 14:567. [PMID: 34742326 PMCID: PMC8572477 DOI: 10.1186/s13071-021-05075-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mast cells are known to affect the primary and secondary immune responses against parasites, and this effect is partially mediated through the release of pro-angiogenic mediators. The aim of this study was to explore the effect of the mast cell stabilizer (MCS), ketotifen, with and without albendazole, an anti-parasitic prescription medicine, on the inflammatory response against Trichinella spiralis, with the overall aim to investigate its effect on angiogenesis accompanying nurse cell formation. METHODS The effect of ketotifen and albendazole was explored in eight groups of female BALB/c mice. Four groups were sensitized with a small dose of T. spiralis larvae. The drug regimen was then applied to both sensitized (challenged) and non-sensitized mice. The parasite load was assessed by histopathological examination of the small intestine and muscle tissue, and angiogenesis was assessed by immunohistochemistry to determine the expression of vascular endothelial growth factor (VEGF). RESULTS Sensitized mice showed a significantly lower parasite load and a more pronounced inflammatory response than mice receiving a single infective dose of T. spiralis larvae. All treated groups showed a significant reduction in parasite count compared to the control groups (groups IAa and IBa), reaching approximately an 98.8% reduction in adult parasite count in the sensitized group treated with albendazole (groups IIAb and IIBb). MCS significantly decreased the parasite count during both the intestinal or muscular phases, reduced tissue inflammation, and decreased local VEGF expression, both in the non-sensitized and sensitized groups. CONCLUSION Sensitization with a low dose of T. spiralis larvae was found to confer a partial protective immunity against re-infection and to positively affect the study outcomes, thus underlining the importance of vaccination, but after extensive studies. The anti-angiogenic effect of MCS protects against larval encystation during the muscle phase. The anti-angiogenic potential of albendazole suggests that the action of this anti-helminthic during trichinellosis is not confined to structural damage to the parasite cuticle but includes an effect on host immunopathological response.
Collapse
Affiliation(s)
- Marwa A El-Dardiry
- Department of Medical Parasitology, Faculty of Medicine, Fayoum University, Fayoum, Egypt.
| | - Amany A Abdel-Aal
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Giza, Egypt.,Department of Postgraduate Studies & Scientific Research, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Magda S A Abdeltawab
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Mona El-Sherbini
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Marwa A Hassan
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Asmaa A Abdel-Aal
- Department of Clinical Pathology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Manal Badawi
- Department of Pathology, National Research Center, Giza, Egypt
| | - Shady E Anis
- Department of Pathology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Bahaa-Eldin A Khaled
- Department of Anatomy & Embryology, Faculty of Medicine, Cairo University, Giza, Egypt.,Department of Anatomy, Jouf University, Sakaka, Saudi Arabia
| | - Abeer S Al-Antably
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
6
|
Kim U, Shin C, Kim CY, Ryu B, Kim J, Bang J, Park JH. Albendazole exerts antiproliferative effects on prostate cancer cells by inducing reactive oxygen species generation. Oncol Lett 2021; 21:395. [PMID: 33777218 PMCID: PMC7988661 DOI: 10.3892/ol.2021.12656] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
Benzimidazole derivatives are used for their antihelmintic properties, but have also been reported to exert anticancer effects. In the present study, the anticancer effects of albendazole on prostate cancer cells were assessed using proliferation, clonogenic and migration assays. To investigate the anticancer mechanisms of albendazole, reactive oxygen species (ROS) levels were measured, and the expression of genes associated with oxidative stress and Wnt/β-catenin signaling was confirmed by reverse transcription-quantitative PCR and western blotting. Albendazole selectively inhibited the proliferation of the PC3, DU145, LNCaP and AT2 prostate cancer cell lines at concentrations that did not affect the proliferation of a normal prostate cell line (RWPE-1). Albendazole also inhibited the colony formation and migration of PC3 and DU145 cells, as well as inducing ROS production. Diphenyleneiodonium chloride, an inhibitor of NADPH oxidase (NOX), one of the sources of ROS, decreased basal ROS levels in the PC3 and DU145 cells, but did not reduce albendazole-associated ROS production, suggesting that ROS production following albendazole treatment was NOX-independent. The anticancer effect was decreased when albendazole-induced ROS was reduced by treatment with antioxidants (glutathione and N-acetylcysteine). Furthermore, albendazole decreased the mRNA expression of CDGSH iron sulfur domain 2, which regulates antioxidant activity against ROS, as well as the antioxidant enzymes catalase, and glutathione peroxidase 1 and 3. Albendazole also decreased the mRNA expression of catenin β1 and transcription factor 4, which regulate Wnt/β-catenin signaling and its associated targets, Twist family BHLH transcription factor 1 and BCL2. The albendazole-related decrease in the expression levels of oxidative stress-related genes and Wnt/β-catenin signaling proteins was thought to be associated with ROS production. These results suggest that the antihelmintic drug, albendazole, has inhibitory effects against prostate cancer cells in vitro. Therefore, albendazole may potentially be used as a novel anticancer agent for prostate cancer.
Collapse
Affiliation(s)
- Ukjin Kim
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Changsoo Shin
- Department of Energy Resources Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - C-Yoon Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Bokyeong Ryu
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin Kim
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Junpil Bang
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae-Hak Park
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
7
|
Gu J, Zhang Q, Geng M, Wang W, Yang J, Khan AUR, Du H, Sha Z, Zhou X, He C. Construction of nanofibrous scaffolds with interconnected perfusable microchannel networks for engineering of vascularized bone tissue. Bioact Mater 2021; 6:3254-3268. [PMID: 33778203 PMCID: PMC7970223 DOI: 10.1016/j.bioactmat.2021.02.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Vascularization and bone regeneration are two closely related processes during bone reconstruction. A three-dimensional (3D) scaffold with porous architecture provides a suitable microenvironment for vascular growth and bone formation. Here, we present a simple and general strategy to construct a nanofibrous poly(l-lactide)/poly(ε-caprolactone) (PLLA/PCL) scaffold with interconnected perfusable microchannel networks (IPMs) based on 3D printing technology by combining the phase separation and sacrificial template methods. The regular and customizable microchannel patterns within the scaffolds (spacings: 0.4 mm, 0.5 mm, and 0.6 mm; diameters: 0.8 mm, 1 mm, and 1.2 mm) were made to investigate the effect of microchannel structure on angiogenesis and osteogenesis. The results of subcutaneous embedding experiment showed that 0.5/0.8-IPMs (spacing/diameter = 0.5/0.8) and 0.5/1-IPMs (spacing/diameter = 0.5/1) scaffolds exhibited more vascular network formation as compared with other counterparts. After loading with vascular endothelial growth factor (VEGF), VEGF@IPMs-0.5/0.8 scaffold prompted better human umbilical vein endothelial cells (HUVECs) migration and neo-blood vessel formation, as determined by Transwell migration, scratch wound healing, and chorioallantoic membrane (CAM) assays. Furthermore, the microangiography and rat cranial bone defects experiments demonstrated that VEGF@IPMs-0.5/0.8 scaffold exhibited better performance in vascular network formation and new bone formation compared to VEGF@IPMs-0.5/1 scaffold. In summary, our results suggested that the microchannel structure within the scaffolds could be tailored by an adjustable caramel-based template strategy, and the combination of interconnected perfusion microchannel networks and angiogenic factors could significantly enhance vascularization and bone regeneration. 3D-printed sacrificial templates are used to construct the scaffold with interconnected perfusable microchannel networks. The microchannel structure within scaffolds can be tailored by changing the template specifications. The introduction of VEGF in the microchannel of scaffold promotes the vascular network formation. Microchannel structure and angiogenic factor within scaffold significantly enhance vascularization and bone regeneration.
Collapse
Affiliation(s)
- Jiani Gu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Qianqian Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Mengru Geng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Weizhong Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Jin Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Atta Ur Rehman Khan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Haibo Du
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Zhou Sha
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| |
Collapse
|
8
|
Yang PK, Chou CH, Huang CC, Wen WF, Chen HF, Shun CT, Ho HN, Chen MJ. Obesity alters ovarian folliculogenesis through disrupted angiogenesis from increased IL-10 production. Mol Metab 2021; 49:101189. [PMID: 33592337 PMCID: PMC7933796 DOI: 10.1016/j.molmet.2021.101189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/28/2022] Open
Abstract
Objective Obesity has been reported to have a modulatory effect on the ovulatory functions of patients with polycystic ovary syndrome. The role of adipokines in this obesity-associated ovulatory disturbance has not been extensively explored. In this study, the relationships between obesity, adipokine production from visceral fat, and ovarian folliculogenesis were explored in a mice model of induced obesity. Methods Obesity was induced in female C57BL/6 mice fed ad libitum with high-fat feed and fructose water for 4 weeks. Follicular developments in the ovaries were assessed by histopathology in these diet-induced obese mice. Changes in adipokine expression in the peri-ovarian adipose tissues were screened with an adipokine array. The adipokine with the most significant increase over time was identified. The functions of the adipokine in angiogenic processes were evaluated in a cell model of endothelial proliferation. The in vivo effects of neutralizing this adipokine using specific antibodies were assessed in the same obesity model. Results A high-fat and fructose diet induced an accumulation of early ovarian follicles and a reduction in mature follicles and corpus lutea. The number of microvessels in the early follicles also decreased. The adipokine protein array of the peri-ovarian adipose tissues identified a progressive increase in IL-10 expression with the duration of the obesogenic diet. In vitro experiments in the endothelial cell model confirmed IL-10 as a disrupter of VEGF-induced angiogenesis. Administration of anti-IL-10 antibodies prevented the histopathological changes induced by the obesogenic diet and further highlighted the role of IL-10 in disrupting folliculogenesis. Conclusions Obesity may disrupt normal folliculogenesis through increased production of IL-10 in visceral fats. This relationship may help clarify the reported association between obesity and ovulatory dysfunction, which has been found in patients with polycystic ovary syndrome. However, the duration of this study was short, which limited conclusions on the long-term reproductive outcomes. Obesity increases IL-10 expression in visceral adipose. IL-10 disrupts VEGF-induced angiogenesis in an endothelial cell model. Disrupted angiogenesis is associated with disturbed folliculogenesis. Anti-IL-10 antibody prevents the altered folliculogenesis induced by obesity. Abnormal production of IL-10 may be a cause of dysovulation in obese individuals.
Collapse
Affiliation(s)
- Po-Kai Yang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chu-Chun Huang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Fen Wen
- Department of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Fu Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Tung Shun
- Department of Forensic Medicine and Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hong-Nerng Ho
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan; College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan; Livia Shang Yu Wan Chair Professor of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
9
|
Xiong B, Nie Y, Yu Y, Wang S, Zuo X. Reduced miR-16 levels are associated with VEGF upregulation in high-risk myelodysplastic syndromes. J Cancer 2021; 12:1967-1977. [PMID: 33753995 PMCID: PMC7974534 DOI: 10.7150/jca.52455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/26/2020] [Indexed: 11/05/2022] Open
Abstract
Objective: Overexpression of vascular endothelial growth factor (VEGF), a major angiogenic factor, was found in myelodysplastic syndromes (MDS) and showed different expression statuses in different risk groups of MDS. We aimed to investigate the possible role of microRNA (miR)-15a and miR-16 on the regulation of VEGF expression and their effect on angiogenesis in lower- and higher-risk MDS. Methods: We studied peripheral blood and bone marrow samples of MDS patients or several leukaemia and MDS cell lines by enzyme-linked immunosorbent assay, immunohistochemical staining, immunofluorescence and quantitative PCR for expression levels of VEGF, miR-15a and miR-16. MiRNA transfection and Luciferase reporter assays were conducted to investigate whether VEGF is a target of miR-16. Migration and tube formation assays were performed in cells exposed to medium from cells with overexpressed or knockdown miR-16. Results: It showed a significantly lower level of miR-16 in higher-risk MDS patients, while the VEGF levels were upregulated. Inverse correlation between VEGF and miR-16 were determined in cells lines including SKM-1, THP-1, and K562 cells. Overexpression of miR-16 in SKM-1 cells resulted in reduced VEGF secretion and cell protein levels. Direct binding of miR-16 to the 3' untranslated region (3'-UTR) of VEGF was confirmed by luciferase reporter assays. The migration and tube formation of human umbilical vein endothelial cells decreased in the presence of medium from SKM-1 cells with overexpressed miR-16. Conclusion: These data suggest that miR-16 may play a role in angiogenesis in higher-risk MDS by targeting VEGF and therefore modulating MDS progression. MiR-16 might be a novel therapeutic target in higher-risk MDS.
Collapse
Affiliation(s)
- Bei Xiong
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | | | - Yalan Yu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shixuan Wang
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuelan Zuo
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Zhang RH, Wang S, Zhang H, Lan JJ, Xu GB, Zhao YL, Wang L, Li YJ, Wang YL, Zhou YH, Liu JL, Pan WD, Liao SG, Zhou M. Discovery of tetrandrine derivatives as tumor migration, invasion and angiogenesis inhibitors. Bioorg Chem 2020; 101:104025. [DOI: 10.1016/j.bioorg.2020.104025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/07/2020] [Accepted: 06/12/2020] [Indexed: 12/21/2022]
|
11
|
Movahedi F, Wu Y, Gu W, Xu ZP. Nanostructuring a Widely Used Antiworm Drug into the Lipid-Coated Calcium Phosphate Matrix for Enhanced Skin Tumor Treatment. ACS APPLIED BIO MATERIALS 2020; 3:4230-4238. [DOI: 10.1021/acsabm.0c00313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Fatemeh Movahedi
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia
| | - Yilun Wu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
12
|
Majnooni MB, Fakhri S, Smeriglio A, Trombetta D, Croley CR, Bhattacharyya P, Sobarzo-Sánchez E, Farzaei MH, Bishayee A. Antiangiogenic Effects of Coumarins against Cancer: From Chemistry to Medicine. Molecules 2019; 24:molecules24234278. [PMID: 31771270 PMCID: PMC6930449 DOI: 10.3390/molecules24234278] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis, the process of formation and recruitment of new blood vessels from pre-existing vessels, plays an important role in the development of cancer. Therefore, the use of antiangiogenic agents is one of the most critical strategies for the treatment of cancer. In addition, the complexity of cancer pathogenicity raises the need for multi-targeting agents. Coumarins are multi-targeting natural agents belonging to the class of benzopyrones. Coumarins have several biological and pharmacological effects, including antimicrobial, antioxidant, anti-inflammation, anticoagulant, anxiolytic, analgesic, and anticancer properties. Several reports have shown that the anticancer effect of coumarins and their derivatives are mediated through targeting angiogenesis by modulating the functions of vascular endothelial growth factor as well as vascular endothelial growth factor receptor 2, which are involved in cancer pathogenesis. In the present review, we focus on the antiangiogenic effects of coumarins and related structure-activity relationships with particular emphasis on cancer.
Collapse
Affiliation(s)
- Mohammad Bagher Majnooni
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran;
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Palatucci, 98168 Messina, Italy; (A.S.); (D.T.)
| | - Domenico Trombetta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Palatucci, 98168 Messina, Italy; (A.S.); (D.T.)
| | | | - Piyali Bhattacharyya
- Escuela de Ciencias de la Salud, Universidad Ana G. Méndez, Recinto de Gurabo, Gurabo, PR 00778, USA;
| | - Eduardo Sobarzo-Sánchez
- Laboratory of Pharmaceutical Chemistry, Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; or
- Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8330507, Chile
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
- Correspondence: (M.H.F.); or (A.B.)
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
- Correspondence: (M.H.F.); or (A.B.)
| |
Collapse
|
13
|
Fakhri S, Abbaszadeh F, Jorjani M, Pourgholami MH. The effects of anticancer medicinal herbs on vascular endothelial growth factor based on pharmacological aspects: a review study. Nutr Cancer 2019; 73:1-15. [DOI: 10.1080/01635581.2019.1673451] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Jorjani
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
14
|
Liu B, Lan M, Wei H, Zhang D, Liu J, Teng J. Downregulated microRNA‑133a induces HUVECs injury: Potential role of the (pro) renin receptor in angiotensin II‑dependent hypertension. Mol Med Rep 2019; 20:2796-2804. [PMID: 31524252 PMCID: PMC6691251 DOI: 10.3892/mmr.2019.10519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/12/2019] [Indexed: 01/17/2023] Open
Abstract
The renin‑angiotensin system (RAS) serves an essential role in hypertension. MicroRNAs (miRs) have been reported to be important regulators in angiotensin (Ang) II‑dependent hypertension. We aimed to explore the roles of Ang II and miR‑133a in the mechanism underlying hypertension. Human umbilical vein endothelial cells (HUVECs) were identified by immunofluorescence staining. Cell viability and miR‑133a expression under the inhibition of Ang II of various concentrations were determined by an MTT assay and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR), respectively. The effects of HUVECs transfected with miR‑133a mimic or inhibitor on Ang II‑induced apoptosis were measured using flow cytometry. The potential targeting of miR‑133a to the 3' untranslated region of (pro) renin receptor (PRR) was assessed using TargetScan and a dual‑luciferase assay. The effects of PRR interference using small interfering (si)RNA on PRR expression and the rate of apoptosis were determined by RT‑qPCR, western blotting and flow cytometry, respectively. Ang II at a concentration of 10‑5 M significantly inhibited the cell viability (P<0.05) and miR‑133a expression (P<0.01); Downregulation of miR‑133a suppressed cell viability. HUVECs transfected with miR‑133a mimic reduced the rate of Ang II‑induced apoptosis from 21.99 to 12.38%, but miR‑133a inhibitor promoted Ang II‑induced apoptosis (apoptosis rate, 28.9%). PRR was predicted to be a target gene of miR‑133a. Transfection with siPRR decreased the apoptotic rate in Ang II + negative control and Ang II + miR‑133a inhibitor group to 11.39 and 12.94%, respectively. Our findings also suggested that Ang II promoted PRR expression to enhance the apoptotic rate of HUVECs via the suppression of miR‑133a. Furthermore, siPRR efficiently decreased the Ang II‑induced apoptosis.
Collapse
Affiliation(s)
- Bing Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology of China, Beijing 100730, P.R. China
| | - Ming Lan
- Department of Cardiology, Beijing Hospital, National Center of Gerontology of China, Beijing 100730, P.R. China
| | - Huali Wei
- Department of Gynecology and Obstetrics, China Meitan General Hospital, Beijing 100028, P.R. China
| | - Dapeng Zhang
- Heart Center and Beijing Key Laboratory of Hypertension Disease, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100028, P.R. China
| | - Junmeng Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology of China, Beijing 100730, P.R. China
| | - Jiwei Teng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
15
|
Benzimidazoles Downregulate Mdm2 and MdmX and Activate p53 in MdmX Overexpressing Tumor Cells. Molecules 2019; 24:molecules24112152. [PMID: 31181622 PMCID: PMC6600429 DOI: 10.3390/molecules24112152] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor suppressor p53 is mutated in about 50% of cancers. Most malignant melanomas carry wild-type p53, but p53 activity is often inhibited due to overexpression of its negative regulators Mdm2 or MdmX. We performed high throughput screening of 2448 compounds on A375 cells carrying p53 activity luciferase reporter construct to reveal compounds that promote p53 activity in melanoma. Albendazole and fenbendazole, two approved and commonly used benzimidazole anthelmintics, stimulated p53 activity and were selected for further studies. The protein levels of p53 and p21 increased upon the treatment with albendazole and fenbendazole, indicating activation of the p53–p21 pathway, while the levels of Mdm2 and MdmX decreased in melanoma and breast cancer cells overexpressing these proteins. We also observed a reduction of cell viability and changes of cellular morphology corresponding to mitotic catastrophe, i.e., G2/M cell cycle arrest of large multinucleated cells with disrupted microtubules. In summary, we established a new tool for testing the impact of small molecule compounds on the activity of p53 and used it to identify the action of benzimidazoles in melanoma cells. The drugs promoted the stability and transcriptional activity of wild-type p53 via downregulation of its negative regulators Mdm2 and MdmX in cells overexpressing these proteins. The results indicate the potential for repurposing the benzimidazole anthelmintics for the treatment of cancers overexpressing p53 negative regulators.
Collapse
|
16
|
Li Q, Cheng K, Wang AY, Xu QG, Fu ZF, He SY, Xu PX. microRNA-126 inhibits tube formation of HUVECs by interacting with EGFL7 and down-regulating PI3K/AKT signaling pathway. Biomed Pharmacother 2019; 116:109007. [PMID: 31170663 DOI: 10.1016/j.biopha.2019.109007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 12/28/2022] Open
Abstract
It's critical for tube formation and angiogenesis to repair ischemic myocardium or stroke. This study aimed to investigate role of microRNA-126 (miR-126) in tube formation in human umbilical vein endothelial cells (HUVECs) and associated mechanisms. Primary neural stem cells (NSCs) and HUVECs were cultured and transfected with microRNA-126 mimics and miR-126 inhibitor. Cell counting kit-8 (CCK-8) and cell cycle assay were conducted for evaluating NSCs viability. Transwell assay was conducted to observe invasive ability of HUVECs. Quantitative real-time PCR (qRT-PCR) assay was used to examine epidermal growth factor like domain 7 (EGFL7) and miR-126 mRNA both in vitro and animal models. Tube forming capability was evaluated in HUVECs. Dual luciferase assay was performed to evaluate interaction between miR-126 and EGFL7 gene. Western blot assay was used to determine phosphoinositide-3-kinase/protein kinase-B (PI3K/AKT) signaling molecules and EGFL7. The results indicated that miR-126 significantly decreased cell viability, inhibited invasive ability and modulated cell cycle of NSCs compared to miR-NC group (p < 0.05). miR-126 significantly inhibited tube formation of HUVECs compared to miR-NC group (p < 0.05). miR-126 significantly down-regulated EGFL7 mRNA and protein expression compared to miR-NC (p < 0.05). Atorvastatin significantly increased CD34 and enhanced EGFL7 expression in traumatic brain injury (TBI) rats brain tissues compared to Model group (p < 0.05). miR-126 significantly down-regulated and atorvastatin up-regulated PI3K/AKT signaling pathway (p < 0.05). Atorvastatin significantly increased EGFL7 and down-regulated miR-126 expression in TBI rats brain tissues compared to Model group (p < 0.05). miR-126 interacted with and negatively correlated with EGFL7 gene both in vitro and in TBI models. In conclusion, microRNA-126 inhibited tube formation of HUVECs by interacting with EGFL7 and down-regulating PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Qiang Li
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China.
| | - Kai Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ai-Yue Wang
- Department of Neurology, Haikou Municipal People's Hospital, Haikou, China
| | - Qiong-Guang Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhou-Feng Fu
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shao-Yu He
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Peng-Xiang Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
17
|
Magnetic solid lipid nanoparticles co-loaded with albendazole as an anti-parasitic drug: Sonochemical preparation, characterization, and in vitro drug release. J Mol Liq 2018. [DOI: 10.1016/j.molliq.2018.06.116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
18
|
Chen S, Ye ZQ, Li ZW, Zhao CX, Chen GJ, Zhou JZ, Wang C, Huang RL, Hong YD. Wenyang Huoxue Jiedu formula inhibits thin-cap fibroatheroma plaque formation via the VEGF/VEGFR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2018; 219:213-221. [PMID: 29551453 DOI: 10.1016/j.jep.2018.03.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 07/09/2017] [Accepted: 03/14/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE For many years, Guangzhou University of Chinese Medicine has been successfully using the empirical Wenyang Huoxue Jiedu formula (WHJF) to treat coronary heart disease. Modern theories of acute coronary syndrome mainly focus on rupture of thin-cap fibroatheromas (TCFAs), which is closely related to the release of vascular endothelial growth factor and its receptor (VEGF/VEGFR). AIM OF STUDY We investigated the effects of WHJF on the formation of TCFA plaques and the potential mechanism (VEGF/VEGFR signaling pathway). MATERIALS AND METHODS For the in vivo experiments, WHJF was administered to ApoE-/- mice, as a model of TCFA plaque formation. Aortic sections of the mice were obtained, and the vulnerability index and new vessel density of plaques were calculated by the Movat staining assay and immunohistochemistry kit, respectively. Protein and mRNA expression levels of VEGF/VEGFR in aortas were assayed by capillary electrophoresis immunoassay and quantitative real-time polymerase chain reaction analyses. In vitro, WHJF serum was produced in rats on the fourth day 2 h after the first administration of different concentrations of WHJF. Proliferation, migration, and lumen formation ability of human umbilical vein endothelial cells (HUVECs) treated with sera from these rats were assayed by the CKK-8 kit, Transwell plates, and Matrigel assay, respectively. Protein and mRNA expression levels of signaling molecules in the VEGF/VEGFR pathways were also examined. RESULTS In vivo, the vulnerability index and new vessel density of plaques in the WHJF group were lower than those values in the blank control group (P < 0.05). No differences were found between the groups in the expression levels of VEGF/VEGFR (P > 0.05). In vitro, the proliferation, migration, and tube formation of HUVECs in the high-dose WHJF group were reduced compared to the control group (P < 0.05). This finding was in agreement with the downregulation of VEGFR-2 and pERK (P < 0.05). The mRNA expression of signaling molecules showed no difference between the groups (P > 0.05). CONCLUSIONS WHJF inhibits TCFA formation by influencing the VEGF/VEGFR signaling pathway.
Collapse
MESH Headings
- Animals
- Drugs, Chinese Herbal/isolation & purification
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Female
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Male
- Mice
- Mice, Knockout
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Rats
- Rats, Sprague-Dawley
- Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors
- Receptors, Vascular Endothelial Growth Factor/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Shuang Chen
- Guangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou, Guangdong 510800, China
| | - Zi-Qing Ye
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Zhi-Wen Li
- Baoan Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong 518000, China
| | - Chun-Xiao Zhao
- Department of Japanese-Oriental (Kampo) Medicine, Graduate School of Medicine, Chiba University, Chiba 2608076, Japan
| | - Guang-Jin Chen
- Yang Jiang Hospital of Traditional Chinese Medicine, Yangjiang, Guangdong 529500, China
| | - Jun-Zhuo Zhou
- Kai Ping Hospital of Traditional Chinese Medicine, Kaiping, Jiangmen, Guangdong 529300, China
| | - Chuan Wang
- Beijing Fengtai Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Beijing 100000, China
| | - Rui-Li Huang
- Department of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Yong-Dun Hong
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China.
| |
Collapse
|
19
|
Lu H, Noorani L, Jiang Y, Du AW, Stenzel MH. Penetration and drug delivery of albumin nanoparticles into pancreatic multicellular tumor spheroids. J Mater Chem B 2017; 5:9591-9599. [PMID: 32264572 DOI: 10.1039/c7tb02902k] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Albumin-based nanoparticles have been exploited as a useful carrier for the efficient delivery of anti-cancer drugs. In this study, albendazole was encapsulated into bovine serum albumin (BSA)-polycaprolactone (PCL) conjugates and the formed nanoparticles with a size about 100 nm were used to treat pancreatic carcinoma cells. In addition, two more types of albendazole-loaded BSA nanoparticles, 10 nm and 200 nm ones, were prepared using a desolvation method. The albendazole-loaded BSA nanoparticles were evaluated with both 2D cultured AsPC-1 cells and 3D multicellular tumor spheroids (MCTS). Their anti-tumor effects were also compared. BSA-PCL nanoparticles and 200 nm BSA nanoparticles showed noticeable cytotoxicity to 2D cultured AsPC-1 cells when compared to the free drug. The penetration of BSA-PCL nanoparticles and 200 nm BSA nanoparticles, especially the BSA-PCL nanoparticles, enabled effective delivery of albendazole into pancreatic MCTS. BSA-PCL nanoparticles also showed a better inhibition effect on the growth of pancreatic MCTS than the 200 nm counterpart. Although 10 nm BSA nanoparticles inhibited the growth of MCTS, the inhibitory effect was even less than that of free albendazole. In addition, it is also found that SPARC protein facilitates the penetration and drug delivery of albumin nanoparticle since treatment using anti-SPARC antibody decreased the efficacy of drug loaded BSA nanoparticles.
Collapse
Affiliation(s)
- Hongxu Lu
- Centre for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Kensington, Sydney, New South Wales 2052, Australia.
| | | | | | | | | |
Collapse
|
20
|
Feizpour F, Jafarpour M, Rezaeifard A. A Tandem Aerobic Photocatalytic Synthesis of Benzimidazoles by Cobalt Ascorbic Acid Complex Coated on TiO2 Nanoparticles Under Visible Light. Catal Letters 2017. [DOI: 10.1007/s10562-017-2232-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
21
|
Movahedi F, Li L, Gu W, Xu ZP. Nanoformulations of albendazole as effective anticancer and antiparasite agents. Nanomedicine (Lond) 2017; 12:2555-2574. [PMID: 28954575 DOI: 10.2217/nnm-2017-0102] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Initially emerging as a widely used clinical antiparasitic drug, albendazole (ABZ) has been increasingly recognized as an effective anticancer agent due to its outstanding advantage, in other words, low toxicity to normal cells but high effectiveness against parasites and some tumors. The major challenge is its poor water solubility and subsequently low bioavailability. This article thus first reviews the brief achievements in using ABZ to treat parasites and cancers, and summarizes the basic mechanisms of action of ABZ. Then this article critically reviews recent nanotechnological strategies, in other words, formulating/conjugating it with carriers into nanoformulations, in practices of improving aqueous solubility and efficacy in treatment of tumors and parasites. Our expert opinions in this field are provided for more effective delivery of ABZ to treat tumors and parasites in vivo.
Collapse
Affiliation(s)
- Fatemeh Movahedi
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Li Li
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
22
|
Li S, Hsu CW, Sakamuru S, Zou C, Huang R, Xia M. Identification of Angiogenesis Inhibitors Using a Co-culture Cell Model in a High-Content and High-Throughput Screening Platform. SLAS Technol 2017; 23:217-225. [PMID: 28922619 PMCID: PMC6032403 DOI: 10.1177/2472630317729792] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Angiogenesis is an important hallmark of cancer, contributing to tumor formation
and metastasis. In vitro angiogenesis models for analyzing tube formation serve
as useful tools to study these processes. However, current in vitro co-culture
models using primary cells have limitations in usefulness and consistency.
Therefore, in the present study, an in vitro co-culture assay system was
optimized in a 1536-well format for high-throughput screening using human
telomerase reverse transcriptase (hTERT)–immortalized mesenchymal stem cells and
aortic endothelial cells. The National Center for Advancing Translational
Sciences (NCATS) Pharmaceutical Collection (NPC) library containing 2816 drugs
was evaluated using the in vitro co-culture assay. From the screen, 35 potent
inhibitors (IC50 ≤1 µM) were identified, followed by 15 weaker
inhibitors (IC50 1–50 µM). Moreover, many known angiogenesis
inhibitors were identified, such as topotecan, docetaxel, and bortezomib.
Several potential novel angiogenesis inhibitors were also identified from this
study, including thimerosal and podofilox. Among the inhibitors, some compounds
were proved to be involved in the hypoxia-inducible factor-1α (HIF-1α) and the
nuclear factor-kappa B (NF-κB) pathways. The co-culture model developed by using
hTERT-immortalized cell lines described in this report provides a consistent and
robust in vitro system for antiangiogenic drug screening.
Collapse
Affiliation(s)
- Shuaizhang Li
- 1 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Chia-Wen Hsu
- 1 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Srilatha Sakamuru
- 1 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Chaozhong Zou
- 2 American Type Culture Collection, Gaithersburg, MD, USA
| | - Ruili Huang
- 1 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Menghang Xia
- 1 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
23
|
A Critical Analysis of the Available In Vitro and Ex Vivo Methods to Study Retinal Angiogenesis. J Ophthalmol 2017; 2017:3034953. [PMID: 28848677 PMCID: PMC5564124 DOI: 10.1155/2017/3034953] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is a biological process with a central role in retinal diseases. The choice of the ideal method to study angiogenesis, particularly in the retina, remains a problem. Angiogenesis can be assessed through in vitro and in vivo studies. In spite of inherent limitations, in vitro studies are faster, easier to perform and quantify, and typically less expensive and allow the study of isolated angiogenesis steps. We performed a systematic review of PubMed searching for original articles that applied in vitro or ex vivo angiogenic retinal assays until May 2017, presenting the available assays and discussing their applicability, advantages, and disadvantages. Most of the studies evaluated migration, proliferation, and tube formation of endothelial cells in response to inhibitory or stimulatory compounds. Other aspects of angiogenesis were studied by assessing cell permeability, adhesion, or apoptosis, as well as by implementing organotypic models of the retina. Emphasis is placed on how the methods are applied and how they can contribute to retinal angiogenesis comprehension. We also discuss how to choose the best cell culture to implement these methods. When applied together, in vitro and ex vivo studies constitute a powerful tool to improve retinal angiogenesis knowledge. This review provides support for researchers to better select the most suitable protocols in this field.
Collapse
|
24
|
Ghasemi F, Black M, Vizeacoumar F, Pinto N, Ruicci KM, Le CCSH, Lowerison MR, Leong HS, Yoo J, Fung K, MacNeil D, Palma DA, Winquist E, Mymryk JS, Boutros PC, Datti A, Barrett JW, Nichols AC. Repurposing Albendazole: new potential as a chemotherapeutic agent with preferential activity against HPV-negative head and neck squamous cell cancer. Oncotarget 2017; 8:71512-71519. [PMID: 29069723 PMCID: PMC5641066 DOI: 10.18632/oncotarget.17292] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 04/11/2017] [Indexed: 11/25/2022] Open
Abstract
Albendazole is an anti-helminthic drug that has been shown to exhibit anti-cancer properties, however its activity in head and neck squamous cell cancer (HNSCC) was unknown. Using a series of in vitro assays, we assessed the ability of albendazole to inhibit proliferation in 20 HNSCC cell lines across a range of albendazole doses (1 nM–10 μM). Cell lines that responded to treatment were further examined for cell death, inhibition of migration and cell cycle arrest. Thirteen of fourteen human papillomavirus-negative HNSCC cell lines responded to albendazole, with an average IC50 of 152 nM. In contrast, only 3 of 6 human papillomavirus-positive HNSCC cell lines responded. Albendazole treatment resulted in apoptosis, inhibition of cell migration, cell cycle arrest in the G2/M phase and altered tubulin distribution. Normal control cells were not measurably affected by any dose tested. This study indicates that albendazole acts to inhibit the proliferation of human papillomavirus-negative HNSCC cell lines and thus warrants further study as a potential chemotherapeutic agent for patients suffering from head and neck cancer.
Collapse
Affiliation(s)
- Farhad Ghasemi
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada
| | - Morgan Black
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Frederick Vizeacoumar
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Nicole Pinto
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Kara M Ruicci
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Carson Cao Son Huu Le
- Department of Surgery, Schulich School of Medicine, Western University, London, Ontario, Canada.,Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, Ontario, Canada
| | - Matthew R Lowerison
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada
| | - Hon Sing Leong
- Department of Surgery, Schulich School of Medicine, Western University, London, Ontario, Canada.,Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, Ontario, Canada
| | - John Yoo
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Kevin Fung
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Danielle MacNeil
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - David A Palma
- Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Eric Winquist
- Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Joe S Mymryk
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, London Health Sciences Centre, London, Ontario, Canada.,Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Paul C Boutros
- Informatics and Biocomputing Program, Ontario Institute of Cancer Research, MaRS Centre, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Alessandro Datti
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - John W Barrett
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Anthony C Nichols
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada.,Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| |
Collapse
|
25
|
Enhancing the in vitro anticancer activity of albendazole incorporated into chitosan-coated PLGA nanoparticles. Carbohydr Polym 2017; 159:39-47. [DOI: 10.1016/j.carbpol.2016.12.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 10/27/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022]
|
26
|
Kim SH, Kim H, Ku HJ, Park JH, Cha H, Lee S, Lee JH, Park JW. Oxalomalate reduces expression and secretion of vascular endothelial growth factor in the retinal pigment epithelium and inhibits angiogenesis: Implications for age-related macular degeneration. Redox Biol 2016; 10:211-220. [PMID: 27810736 PMCID: PMC5094379 DOI: 10.1016/j.redox.2016.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/20/2016] [Accepted: 10/21/2016] [Indexed: 11/21/2022] Open
Abstract
Clinical and experimental observations indicate a critical role for vascular endothelial growth factor (VEGF), secreted by the retinal pigment epithelium (RPE), in pathological angiogenesis and the development of choroidal neovascularization (CNV) in age-related macular degeneration (AMD). RPE-mediated VEGF expression, leading to angiogenesis, is a major signaling mechanism underlying ocular neovascular disease. Inhibiting this signaling pathway with a therapeutic molecule is a promising anti-angiogenic strategy to treat this disease with potentially fewer side effects. Oxalomalate (OMA) is a competitive inhibitor of NADP+-dependent isocitrate dehydrogenase (IDH), which plays an important role in cellular signaling pathways regulated by reactive oxygen species (ROS). Here, we have investigated the inhibitory effect of OMA on the expression of VEGF, and the associated underlying mechanism of action, using in vitro and in vivo RPE cell models of AMD. We found that OMA reduced the expression and secretion of VEGF in RPE cells, and consequently inhibited CNV formation. This function of OMA was linked to its capacity to activate the pVHL-mediated HIF-1α degradation in these cells, partly via a ROS-dependent ATM signaling axis, through inhibition of IDH enzymes. These findings reveal a novel role for OMA in inhibiting RPE-derived VEGF expression and angiogenesis, and suggest unique therapeutic strategies for treating pathological angiogenesis and AMD development. Oxalomalate reduces VEGF expression in RPE cells by promoting HIF-1α degradation. Oxalomalate activates pVHL-mediated HIF-1α degradation by regulation of ATM-Chk2-E2F1 axis. Inhibition of IDH enzymes by oxalomalate activates ROS-mediated ATM signaling axis. Oxalomalate inhibits CNV-related angiogenesis in in vivo mouse model of AMD.
Collapse
Affiliation(s)
- Sung Hwan Kim
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hyunjin Kim
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hyeong Jun Ku
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jung Hyun Park
- Department of Food and Biotechnology, Korea University, Sejong, Republic of Korea
| | - Hanvit Cha
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Seoyoon Lee
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong, Republic of Korea; Institutes of Natural Sciences, Korea University, Sejong, Republic of Korea.
| | - Jeen-Woo Park
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
27
|
Liu HT, Xing AY, Chen X, Ma RR, Wang YW, Shi DB, Zhang H, Li P, Chen HF, Li YH, Gao P. MicroRNA-27b, microRNA-101 and microRNA-128 inhibit angiogenesis by down-regulating vascular endothelial growth factor C expression in gastric cancers. Oncotarget 2016; 6:37458-70. [PMID: 26460960 PMCID: PMC4741941 DOI: 10.18632/oncotarget.6059] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 09/23/2015] [Indexed: 01/30/2023] Open
Abstract
Vascular Endothelial Growth Factor C (VEGF-C) has critical roles in angiogenesis in human cancers; however, the underlying mechanisms regulating VEGF-C expression remain largely unknown. In the present study, VEGF-C protein expression and the density of blood vessels or lymphatic vessels were determined by immunohistochemistry in 103 cases of gastric cancer tissues. Suppression of VEGF-C by miR-27b, miR-101 and miR-128 was investigated by luciferase assays, Western blot and ELISA. The miRNAs expression levels were detected in human gastric cancers by real-time quantitative PCR. Cell proliferation, migration and invasion assays were performed to assess the effect of miRNAs on gastric cancer cells and human umbilical vascular endothelial cells (HUVECs). Our data showed that high VEGF-C expression was significantly associated with increased tumor size, advanced TNM classification and clinical stage, higher microvessel density (MVD) and lymphatic density (LVD), as well as poor survival in patients with gastric cancer. Furthermore, VEGF-C was found to be a direct target gene of miR-27b, miR-101, and miR-128. The expression levels of the three miRNAs were inversely correlated with MVD. Overexpression of miR-27b, miR-101, or miR-128 suppressed migration, proliferation activity, and tube formation in HUVECs by repressing VEGF-C secretion in gastric cancer cells. We conclude that miR-27b, miR-101 and miR-128 inhibit angiogenesis by down-regulating VEGF-C expression in gastric cancers.
Collapse
Affiliation(s)
- Hai-Ting Liu
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Ai-Yan Xing
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Xu Chen
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Ran-Ran Ma
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Ya-Wen Wang
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Duan-Bo Shi
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Hui Zhang
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Peng Li
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Hong-Fang Chen
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China.,Department of Pathology, Qingzhou Center Hospital, Weifang, P.R. China
| | - Yu-Hong Li
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China.,Department of Pathology, Liaocheng Peoples Hospital, Liaocheng, P.R. China
| | - Peng Gao
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| |
Collapse
|
28
|
Králová V, Hanušová V, Rudolf E, Čáňová K, Skálová L. Flubendazole induces mitotic catastrophe and senescence in colon cancer cells in vitro. J Pharm Pharmacol 2016; 68:208-18. [DOI: 10.1111/jphp.12503] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 10/28/2015] [Indexed: 12/31/2022]
Abstract
Abstract
Objectives
Flubendazole (FLU), a member of benzimidazole family of anthelmintic drugs, is able to inhibit proliferation of various cancer cells. The aim of present study was to elucidate the mechanisms of antiproliferative effect of FLU on colorectal cancer cells in vitro.
Methods
The effect of FLU on proliferation, microtubular network, DNA content, caspase activation and senescence induction was studied in SW480 and SW620 cell lines.
Key findings
Flubendazole significantly affected cell proliferation in a pattern typical for mitotic inhibitor. This was accompanied by decrease in cyclin D1 levels, increase in cyclin B1 levels, activation of caspase 2 and caspase 3/7 and PARP cleavage. Morphological observations revealed disruption of microtubular network, irregular mitotic spindles, formation of giant multinucleated cells and increase in nuclear area and DNA content. In SW620 cell line, 37.5% giant multinucleated cells induced by FLU treatment showed positivity for SA-β-galactosidase staining. Cell lines were able to recover from the treatment and this process was faster in SW480 cells.
Conclusion
Flubendazole in low concentration temporarily inhibits cell proliferation and induces mitotic catastrophe and premature senescence in human colon cancer cells in vitro.
Collapse
Affiliation(s)
- Věra Králová
- Department of Medical Biology and Genetics, Charles University in Prague, Hradec Králové, Czech Republic
| | - Veronika Hanušová
- Department of Medical Biology and Genetics, Charles University in Prague, Hradec Králové, Czech Republic
| | - Emil Rudolf
- Department of Medical Biology and Genetics, Charles University in Prague, Hradec Králové, Czech Republic
| | - Kristýna Čáňová
- Department of Medical Biology and Genetics, Charles University in Prague, Hradec Králové, Czech Republic
| | - Lenka Skálová
- Department of Biochemical Sciences, Charles University in Prague, Hradec Králové, Czech Republic
| |
Collapse
|
29
|
Wu SY, Hou HS, Sun YS, Cheng JY, Lo KY. Correlation between cell migration and reactive oxygen species under electric field stimulation. BIOMICROFLUIDICS 2015; 9:054120. [PMID: 26487906 PMCID: PMC4600077 DOI: 10.1063/1.4932662] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 09/28/2015] [Indexed: 05/04/2023]
Abstract
Cell migration is an essential process involved in the development and maintenance of multicellular organisms. Electric fields (EFs) are one of the many physical and chemical factors known to affect cell migration, a phenomenon termed electrotaxis or galvanotaxis. In this paper, a microfluidics chip was developed to study the migration of cells under different electrical and chemical stimuli. This chip is capable of providing four different strengths of EFs in combination with two different chemicals via one simple set of agar salt bridges and Ag/AgCl electrodes. NIH 3T3 fibroblasts were seeded inside this chip to study their migration and reactive oxygen species (ROS) production in response to different EF strengths and the presence of β-lapachone. We found that both the EF and β-lapachone level increased the cell migration rate and the production of ROS in an EF-strength-dependent manner. A strong linear correlation between the cell migration rate and the amount of intracellular ROS suggests that ROS are an intermediate product by which EF and β-lapachone enhance cell migration. Moreover, an anti-oxidant, α-tocopherol, was found to quench the production of ROS, resulting in a decrease in the migration rate.
Collapse
Affiliation(s)
- Shang-Ying Wu
- Department of Agricultural Chemistry, National Taiwan University , Taipei 10617, Taiwan
| | - Hsien-San Hou
- Research Center for Applied Sciences , Academia Sinica, Taipei 11529, Taiwan
| | - Yung-Shin Sun
- Department of Physics, Fu-Jen Catholic University , New Taipei City 24205, Taiwan
| | - Ji-Yen Cheng
- Research Center for Applied Sciences , Academia Sinica, Taipei 11529, Taiwan
| | - Kai-Yin Lo
- Department of Agricultural Chemistry, National Taiwan University , Taipei 10617, Taiwan
| |
Collapse
|
30
|
Zhao J, Geng YU, Hua H, Cun B, Chen Q, Xi X, Yang L, Li Y. Fenofibrate inhibits the expression of VEGFC and VEGFR-3 in retinal pigmental epithelial cells exposed to hypoxia. Exp Ther Med 2015; 10:1404-1412. [PMID: 26622498 PMCID: PMC4578108 DOI: 10.3892/etm.2015.2697] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 08/20/2015] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to examine the mechanisms through which fenofibrate inhibits the ability of human retinal pigment epithelial cells (RPE cells) exposed to hypoxia to stimulate the proliferation and migration of human umbilical vein endothelial cells (HUVECs). For this purpose, RPE cells and HUVECs were divided into the following groups: RPE-normoxia, RPE + fenofibrate, RPE-hypoxia, RPE hypoxia + fenofibrate; HUVECs normal culture and HUVECs + RPE-hypoxia culture supernatant. RPE cell hypoxia was induced by cobalt(II) chloride (CoCl2). A superoxide anion probe was used to measure the production of superoxide anion, which is indicative of hypoxic conditions. Cell proliferation was assessed by MTT assay, and the expression of vascular endothelial growth factor C (VEGFC) and vascular endothelial growth factor receptor-3 (VEGFR-3) in the RPE cell culture supernatant was measured by enzyme-linked immunosorbent assay (ELISA). The migration ability of the HUVECs was determined by scratch-wound assay, and the angiogenic ability of the HUVECs was examined by measuring cell lumen formation. The mRNA and protein expression levels of VEGFC and VEGFR-3 in the RPE cells were measured by RT-qPCR and western blot analysis, respectively. Our results revealed that fenofibrate inhibited the increase in the expression and release of VEGFC and VEGFR-3 into the RPE cell culture supernatant induced by exposure to hypoxia. The culture of HUVECs in medium supernatant of RPE cells epxosed to hypoxia enhanced the viability and migration ability of the HUVECs and promoted lumen formation; these effects were inhibited by fenofibrate. In conclusion, our data demonstrated that the exposure of RPE cells to hypoxia induced the expression and release of VEGFC and VEGFR-3 into the cell culture supernatant. The culture of HUVECs in conditioned medium from RPE cells exposed to hypoxia increased VEGFC and VEGFR-3 expression, and promoted the proliferation and migration of the HUVECs, as well as capillary tube formation, suggesting that RPE cells play an important role in the formation of choroidal neovascularization resulting from hypoxia. Fenofibrate inhibited the upregulation of VEGFC and VEGFR-3 in the RPE cells exposed to hypoxia, and thus reduced the ability of HUVECs to form new blood vessels.
Collapse
Affiliation(s)
- Jianfeng Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Y U Geng
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Hairong Hua
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Biyun Cun
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Qianbo Chen
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Xiaoting Xi
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Liushu Yang
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Yan Li
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| |
Collapse
|
31
|
Wu F, Song H, Zhang Y, Zhang Y, Mu Q, Jiang M, Wang F, Zhang W, Li L, Li H, Wang Y, Zhang M, Li S, Yang L, Meng Y, Tang D. Irisin Induces Angiogenesis in Human Umbilical Vein Endothelial Cells In Vitro and in Zebrafish Embryos In Vivo via Activation of the ERK Signaling Pathway. PLoS One 2015; 10:e0134662. [PMID: 26241478 PMCID: PMC4524626 DOI: 10.1371/journal.pone.0134662] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/13/2015] [Indexed: 11/18/2022] Open
Abstract
As a link between exercise and metabolism, irisin is assumed to be involved in increased total body energy expenditure, reduced body weight, and increased insulin sensitivity. Although our recent evidence supported the contribution of irisin to vascular endothelial cell (ECs) proliferation and apoptosis, further research of irisin involvement in the angiogenesis of ECs was not conclusive. In the current study, it was found that irisin promoted Human Umbilical Vein Endothelial Cell (HUVEC) angiogenesis via increasing migration and tube formation, and attenuated chemically-induced intersegmental vessel (ISV) angiogenic impairment in transgenic TG (fli1: GFP) zebrafish. It was further demonstrated that expression of matrix metalloproteinase (MMP) 2 and 9 were also up-regulated in endothelial cells. We also found that irisin activated extracellular signal–related kinase (ERK) signaling pathways. Inhibition of ERK signaling by using U0126 decreased the pro-migration and pro-angiogenic effect of irisin on HUVEC. Also, U0126 inhibited the elevated expression of MMP-2 and MMP-9 when they were treated with irisin. In summary, these findings provided direct evidence that irisin may play a pivotal role in maintaining endothelium homeostasis by promoting endothelial cell angiogenesis via the ERK signaling pathway.
Collapse
Affiliation(s)
- Fei Wu
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Haibo Song
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Yuan Zhang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Yuzhu Zhang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Qian Mu
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Miao Jiang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Fang Wang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Wen Zhang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Liang Li
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Huanjie Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, 250012, P.R. China
| | - Yunshan Wang
- Jinan Central Hospital Affiliated to Shandong University, Jinan, 250012, P.R. China
| | - Mingxiang Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Shiwu Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States of America
| | - Lijun Yang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States of America
| | - Yan Meng
- Department of Urology, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
- * E-mail: (DT); (YM)
| | - Dongqi Tang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P.R. China
- * E-mail: (DT); (YM)
| |
Collapse
|
32
|
Noorani L, Stenzel M, Liang R, Pourgholami MH, Morris DL. Albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model. J Nanobiotechnology 2015; 13:25. [PMID: 25890381 PMCID: PMC4409778 DOI: 10.1186/s12951-015-0082-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/02/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The poor prognosis of patients with drug resistant ovarian cancer and the lack of targeted therapy have raised the need for alternative treatments. Albendazole (ABZ) is an anti-parasite compound capable of impairing microtubule formation. We hypothesized that ABZ could be repurposed as a potential anti-angiogenic drug due to its potent inhibition of vascular endothelial growth factor (VEGF) in ovarian cancer with ascites. However, the poor aqueous solubility of ABZ limits its potential for cancer therapy. In this study, we have assembled ABZ with bovine serum albumin into nanoparticles with a size range of 7-10 nm (BSA-ABZ) and 200-250 nm (Nab-ABZ). We further examined the anticancer effects of ABZ carrying nanoparticles in ovarian cancer cells, in both in vitro and in vivo models. RESULTS Drug release studies demonstrated that about 93% of ABZ was released from BSA-ABZ 10 nm in comparison to 83% from Nab-ABZ 200 nm at pH 7.4 in 8 days. In vitro cell proliferation studies showed that the BSA-ABZ 10 nm exhibited the highest killing efficacy of ovarian cancer cells with surprisingly least toxicity to healthy ovarian epithelial cells. Confocal microscopy and fluorescence activated cell sorting analysis (FACS) revealed more efficient internalization of the BSA-ABZ 10 nm by cancer cells. For in vivo studies, we examined the tumor growth, ascites formation and the expression of VEGF and secreted protein acidic and rich in cysteine (SPARC) in tumor samples and only VEGF in plasma samples. The BSA-ABZ 10 nm reduced the tumor burden significantly (p < 0.02) at a much lower drug dose (10 μg/ml) compare to free drug. Both formulations were capable of suppressing the ascites volume significantly (p < 0.05) and reducing the number of ascites cells. The expression of VEGF and SPARC was also reduced, which indicates the underlying therapeutic mechanism of the ABZ. CONCLUSION Our data suggest that the BSA-ABZ may hold promise for the treatment and control of progression of ovarian cancer with ascites. However further studies are required to examine the efficacy of both the formulations in aggressive models of recurrent ovarian cancer with respect to particle size and dosing parameters.
Collapse
Affiliation(s)
- Lubna Noorani
- Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, NSW, Australia.
- Department of Surgery, St. George Clinical School, Faculty of Medicine, University of New South Wales, Kogarah, NSW, Australia.
| | - Martina Stenzel
- Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, NSW, Australia.
| | - Roger Liang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.
| | - Mohammad H Pourgholami
- Department of Surgery, St. George Clinical School, Faculty of Medicine, University of New South Wales, Kogarah, NSW, Australia.
| | - David L Morris
- Department of Surgery, St. George Clinical School, Faculty of Medicine, University of New South Wales, Kogarah, NSW, Australia.
| |
Collapse
|
33
|
Qiao C, Jiang Y, Deng C, Huang Z, Teng K, Chen L, Liu X. Characterization of the transcriptional activation domains of human TEF3-1 (transcription enhancer factor 3 isoform 1). Arch Biochem Biophys 2015; 569:54-61. [PMID: 25687649 DOI: 10.1016/j.abb.2015.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/23/2015] [Accepted: 02/04/2015] [Indexed: 10/24/2022]
Abstract
TEF3-1 (transcription enhancer factor 3 isoform 1) is a human transcriptional factor, which has a N-terminal TEA/ATTS domain supposedly for DNA binding and C-terminal PRD and STY domains for transcriptional activation. Taking advantage of the efficient reporter design of yeast two-hybrid system, we characterized the TEF3-1 domains in activating gene expression. Previously study usually mentioned that the C-terminal domain of TEF3-1 has the transcriptional activity, however, our data shows that the peptides TEF3-11-66 and TEF3-1197-434 functioned as two independent activation domains, suggesting that N-terminal domain of TEF3-1 also has transcriptional activation capacity. Additionally, more deletions of amino acids 197-434 showed that only the peptides TEF3-1197-265 contained the minimum sequences for the C-terminal transcriptional activation domain. The protein structure is predicted to contain a helix-turn-helix structure in TEF3-11-66 and four β sheets in TEF3-1197-265. Finally, after the truncated fragments of TEF3-1 were expressed in HUVEC cells, the whole TEF3-1 and the two activation domains could increase F-actin stress fiber, cell proliferation, migration and targeted gene expression. Further analysis and characterization of the activation domains in TEF3-1 may broaden our understanding of the gene involved in angiogenesis and other pathological processes.
Collapse
Affiliation(s)
- Cheng Qiao
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yajie Jiang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Cuilan Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Zebo Huang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Kaixuan Teng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Lan Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xin Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
34
|
Seyedpour SM, Pachenari M, Janmaleki M, Alizadeh M, Hosseinkhani H. Effects of an antimitotic drug on mechanical behaviours of the cytoskeleton in distinct grades of colon cancer cells. J Biomech 2014; 48:1172-8. [PMID: 25678199 DOI: 10.1016/j.jbiomech.2014.11.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 11/17/2014] [Accepted: 11/20/2014] [Indexed: 11/28/2022]
Abstract
Biomechanical behaviours of cells change during cancer progression due to alterations in the main cytoskeletal proteins. Microtubules play a vital role in mitosis and in supporting the integrity of the cell due to their ability to withstand high compressive loads. Accordingly, microtubule-targeting agents (MTAs) have become one of the most promising classes of drugs in cancer therapy. This study evaluated changes in visco-elastic parameters induced by an appropriate concentration of an antimitotic drug in two different grades of colon cancer cells. Actin microfilaments and microtubules contents in the cells were evaluated by Western blot analysis and fluorescence intensity calculation. Micropipette aspiration experiments showed that the MTA had distinct mechanical effects on different cell lines. The more aggressive the cells, the greater the reduction in elasticity and viscosity. Invasive cells had a higher initial instantaneous Young's modulus than primary cells, but this reduced to approximately one half of the values for primary cells after 48 h of drug treatment. A considerable association was seen between the changes in mechanical properties and the microtubule to F-actin microfilament content ratio, which decreased with MTA treatment.
Collapse
Affiliation(s)
- S M Seyedpour
- Department of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - M Pachenari
- Department of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - M Janmaleki
- Medical Nanotechnology and Tissue Engineering Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - M Alizadeh
- Department of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran
| | - H Hosseinkhani
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| |
Collapse
|
35
|
Gold nanoparticles inhibit VEGF165-induced migration and tube formation of endothelial cells via the Akt pathway. BIOMED RESEARCH INTERNATIONAL 2014; 2014:418624. [PMID: 24987682 PMCID: PMC4058682 DOI: 10.1155/2014/418624] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/27/2014] [Accepted: 04/28/2014] [Indexed: 02/07/2023]
Abstract
The early stages of angiogenesis can be divided into three steps: endothelial cell proliferation, migration, and tube formation. Vascular endothelial growth factor (VEGF) is considered the most important proangiogenic factor; in particular, VEGF165 plays a critical role in angiogenesis. Here, we evaluated whether gold nanoparticles (AuNPs) could inhibit the VEGF165-induced human umbilical vein endothelial cell (HUVEC) migration and tube formation. AuNPs and VEGF165 were coincubated overnight at 4°C, after which the effects on cell migration and tube formation were assessed. Cell migration was assessed using a modified wound-healing assay and a transwell chamber assay; tube formation was assessed using a capillary-like tube formation assay and a chick chorioallantoic membrane (CAM) assay. We additionally detected the cell surface morphology and ultrastructure using atomic force microscopy (AFM). Furthermore, Akt phosphorylation downstream of VEGFR-2/PI3K in HUVECs was determined in a Western blot analysis. Our study demonstrated that AuNPs significantly inhibited VEGF165-induced HUVEC migration and tube formation by affecting the cell surface ultrastructure, cytoskeleton and might have inhibited angiogenesis via the Akt pathway.
Collapse
|
36
|
Bai GY, Lan XW, Chen GF, Liu XF, Li TY, Shi LJ. A novel sodium iodide and ammonium molybdate co-catalytic system for the efficient synthesis of 2-benzimidazoles using hydrogen peroxide under ultrasound irradiation. ULTRASONICS SONOCHEMISTRY 2014; 21:520-526. [PMID: 24074960 DOI: 10.1016/j.ultsonch.2013.08.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/28/2013] [Accepted: 08/28/2013] [Indexed: 06/02/2023]
Abstract
The reaction of aldehydes and o-phenylenediamine for the preparation of 2-benzimidazoles has been studied using hydrogen peroxide as an oxidant under ultrasound irradiation at room temperature in this paper. The combination of substoichiometric sodium iodide and ammonium molybdate as co-catalysts, together with using small amounts of hydrogen peroxide, makes this transformation very efficient and attractive under ultrasound. Thus, a mild, green and efficient method is established to carry out this reaction in high yield.
Collapse
Affiliation(s)
- Guo-Yi Bai
- Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding 071002, PR China.
| | | | | | | | | | | |
Collapse
|
37
|
Temirak A, Shaker YM, Ragab FAF, Ali MM, Soliman SM, Mortier J, Wolber G, Ali HI, Diwani HIE. Synthesis, Biological Evaluation, and Docking Studies of New 2-Furylbenzimidazoles as Anti-Angiogenic Agents: Part II. Arch Pharm (Weinheim) 2014; 347:291-304. [DOI: 10.1002/ardp.201300356] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 11/12/2013] [Accepted: 11/15/2013] [Indexed: 01/28/2023]
Affiliation(s)
- Ahmed Temirak
- Division of Pharmaceutical and Drug Industries, Department of Chemistry of Natural and Microbial Products; National Research Center; Dokki Cairo Egypt
| | - Yasser M. Shaker
- Division of Pharmaceutical and Drug Industries, Department of Chemistry of Natural and Microbial Products; National Research Center; Dokki Cairo Egypt
| | - Fatma A. F. Ragab
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry; Cairo University; Giza Egypt
| | - Mamdouh M. Ali
- Division of Genetic Engineering and Biotechnology, Department of Biochemistry; National Research Center; Cairo Egypt
| | - Salwa M. Soliman
- Department of Pharmaceutical Chemistry; Institute of Pharmacy, Free University of Berlin; Berlin Germany
| | - Jeremie Mortier
- Department of Pharmaceutical Chemistry; Institute of Pharmacy, Free University of Berlin; Berlin Germany
- Department of Organic Chemistry; Free University of Berlin; Berlin Germany
| | - Gerhard Wolber
- Department of Pharmaceutical Chemistry; Institute of Pharmacy, Free University of Berlin; Berlin Germany
| | - Hamed I. Ali
- Department of Pharmaceutical Sciences; Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center; Round Rock TX USA
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry; Helwan University; Cairo Egypt
| | - Hoda I. El Diwani
- Division of Pharmaceutical and Drug Industries, Department of Chemistry of Natural and Microbial Products; National Research Center; Dokki Cairo Egypt
| |
Collapse
|
38
|
Huang CY, Pourgholami MH, Allen BJ. Optimizing radioimmunoconjugate delivery in the treatment of solid tumor. Cancer Treat Rev 2012; 38:854-60. [DOI: 10.1016/j.ctrv.2011.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 02/08/2023]
|
39
|
Dakshanamurthy S, Issa NT, Assefnia S, Seshasayee A, Peters OJ, Madhavan S, Uren A, Brown ML, Byers SW. Predicting new indications for approved drugs using a proteochemometric method. J Med Chem 2012; 55:6832-48. [PMID: 22780961 PMCID: PMC3419493 DOI: 10.1021/jm300576q] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The most effective way to move from target identification to the clinic is to identify already approved drugs with the potential for activating or inhibiting unintended targets (repurposing or repositioning). This is usually achieved by high throughput chemical screening, transcriptome matching, or simple in silico ligand docking. We now describe a novel rapid computational proteochemometric method called "train, match, fit, streamline" (TMFS) to map new drug-target interaction space and predict new uses. The TMFS method combines shape, topology, and chemical signatures, including docking score and functional contact points of the ligand, to predict potential drug-target interactions with remarkable accuracy. Using the TMFS method, we performed extensive molecular fit computations on 3671 FDA approved drugs across 2335 human protein crystal structures. The TMFS method predicts drug-target associations with 91% accuracy for the majority of drugs. Over 58% of the known best ligands for each target were correctly predicted as top ranked, followed by 66%, 76%, 84%, and 91% for agents ranked in the top 10, 20, 30, and 40, respectively, out of all 3671 drugs. Drugs ranked in the top 1-40 that have not been experimentally validated for a particular target now become candidates for repositioning. Furthermore, we used the TMFS method to discover that mebendazole, an antiparasitic with recently discovered and unexpected anticancer properties, has the structural potential to inhibit VEGFR2. We confirmed experimentally that mebendazole inhibits VEGFR2 kinase activity and angiogenesis at doses comparable with its known effects on hookworm. TMFS also predicted, and was confirmed with surface plasmon resonance, that dimethyl celecoxib and the anti-inflammatory agent celecoxib can bind cadherin-11, an adhesion molecule important in rheumatoid arthritis and poor prognosis malignancies for which no targeted therapies exist. We anticipate that expanding our TMFS method to the >27 000 clinically active agents available worldwide across all targets will be most useful in the repositioning of existing drugs for new therapeutic targets.
Collapse
Affiliation(s)
- Sivanesan Dakshanamurthy
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center , Washington, DC 20057, United States.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kim Y, Pourgholami MH, Morris DL, Stenzel MH. Effect of Cross-Linking on the Performance of Micelles As Drug Delivery Carriers: A Cell Uptake Study. Biomacromolecules 2012; 13:814-25. [PMID: 22276949 DOI: 10.1021/bm201730p] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Yoseop Kim
- Centre
for Advanced Macromolecular Design (CAMD) and ‡Cancer Research Laboratories, Department
of Surgery, St. George Hospital, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mohammad H. Pourgholami
- Centre
for Advanced Macromolecular Design (CAMD) and ‡Cancer Research Laboratories, Department
of Surgery, St. George Hospital, University of New South Wales, Sydney, NSW 2052, Australia
| | - David L. Morris
- Centre
for Advanced Macromolecular Design (CAMD) and ‡Cancer Research Laboratories, Department
of Surgery, St. George Hospital, University of New South Wales, Sydney, NSW 2052, Australia
| | - Martina H. Stenzel
- Centre
for Advanced Macromolecular Design (CAMD) and ‡Cancer Research Laboratories, Department
of Surgery, St. George Hospital, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
41
|
Amini A, Masoumi Moghaddam S, Morris DL, Pourgholami MH. Utility of vascular endothelial growth factor inhibitors in the treatment of ovarian cancer: from concept to application. JOURNAL OF ONCOLOGY 2011; 2012:540791. [PMID: 21961001 PMCID: PMC3180777 DOI: 10.1155/2012/540791] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 05/26/2011] [Accepted: 05/26/2011] [Indexed: 12/20/2022]
Abstract
Despite recent advances in the management of ovarian cancer, it remains the most lethal gynecologic malignancy. Vascular endothelial growth factor (VEGF) has been shown to play a pivotal role in the progression of ovarian cancer leading to the eventual development of malignant ascites. On this basis, agents rendering VEGF ineffective by neutralizing VEGF (bevacizumab), blocking its receptors (aflibercept), or interfering with the postreceptor signaling pathways (sunitinib) provide us with the rational treatment options. These agents are generally used in combination with the standard chemotherapeutic drugs. Here, we discuss the basis of and the logic behind the use of these agents in the treatment of epithelial ovarian cancer, as well as their evaluation in different preclinical and clinical studies.
Collapse
Affiliation(s)
- Afshin Amini
- Cancer Research Laboratories, Department of Surgery, St George Hospital (SESIAHS), The University of New South Wales, Sydney, NSW 2217, Australia
| | - Samar Masoumi Moghaddam
- Cancer Research Laboratories, Department of Surgery, St George Hospital (SESIAHS), The University of New South Wales, Sydney, NSW 2217, Australia
| | - David L. Morris
- Department of Surgery, St George Hospital (SESIAHS), The University of New South Wales, Sydney, NSW 2217, Australia
| | - Mohammad H. Pourgholami
- Cancer Research Laboratories, Department of Surgery, St George Hospital (SESIAHS), The University of New South Wales, Sydney, NSW 2217, Australia
| |
Collapse
|
42
|
Seaman ME, Peirce SM, Kelly K. Rapid analysis of vessel elements (RAVE): a tool for studying physiologic, pathologic and tumor angiogenesis. PLoS One 2011; 6:e20807. [PMID: 21694777 PMCID: PMC3111429 DOI: 10.1371/journal.pone.0020807] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 05/10/2011] [Indexed: 11/26/2022] Open
Abstract
Quantification of microvascular network structure is important in a myriad of emerging research fields including microvessel remodeling in response to ischemia and drug therapy, tumor angiogenesis, and retinopathy. To mitigate analyst-specific variation in measurements and to ensure that measurements represent actual changes in vessel network structure and morphology, a reliable and automatic tool for quantifying microvascular network architecture is needed. Moreover, an analysis tool capable of acquiring and processing large data sets will facilitate advanced computational analysis and simulation of microvascular growth and remodeling processes and enable more high throughput discovery. To this end, we have produced an automatic and rapid vessel detection and quantification system using a MATLAB graphical user interface (GUI) that vastly reduces time spent on analysis and greatly increases repeatability. Analysis yields numerical measures of vessel volume fraction, vessel length density, fractal dimension (a measure of tortuosity), and radii of murine vascular networks. Because our GUI is open sourced to all, it can be easily modified to measure parameters such as percent coverage of non-endothelial cells, number of loops in a vascular bed, amount of perfusion and two-dimensional branch angle. Importantly, the GUI is compatible with standard fluorescent staining and imaging protocols, but also has utility analyzing brightfield vascular images, obtained, for example, in dorsal skinfold chambers. A manually measured image can be typically completed in 20 minutes to 1 hour. In stark comparison, using our GUI, image analysis time is reduced to around 1 minute. This drastic reduction in analysis time coupled with increased repeatability makes this tool valuable for all vessel research especially those requiring rapid and reproducible results, such as anti-angiogenic drug screening.
Collapse
Affiliation(s)
- Marc E. Seaman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Kimberly Kelly
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|