1
|
Guo S, Zheng X, Chen W, Raza U, Zeng A, Akter F, Huang Q, Yao S. From bench to bedside: Advancing towards therapeutic treatment of vestibular schwannomas. Neurooncol Adv 2024; 6:vdae107. [PMID: 39022647 PMCID: PMC11252569 DOI: 10.1093/noajnl/vdae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024] Open
Abstract
Vestibular schwannomas are rare intracranial tumors originating from Schwann cells of the vestibular nerve. Despite their benign nature, these tumors can exert significant mass effects and debilitating symptoms, including gradual hearing loss, vertigo, facial nerve dysfunction, and headaches. Current clinical management options encompass wait-and-scan, surgery, radiation therapy, and off-label medication. However, each approach exhibits its own challenges and harbors limitations that underscore the urgent need for therapeutic treatments. Over the past 2 decades, extensive elucidation of the molecular underpinnings of vestibular schwannomas has unraveled genetic anomalies, dysregulated signaling pathways, downstream of receptor tyrosine kinases, disrupted extracellular matrix, inflammatory tumor microenvironment, and altered cerebrospinal fluid composition as integral factors in driving the development and progression of the disease. Armed with this knowledge, novel therapeutic interventions tailored to the unique molecular characteristics of those conditions are actively being pursued. This review underscores the urgency of addressing the dearth of Food and Drug Administration-approved drugs for vestibular schwannoma, highlighting the key molecular discoveries and their potential translation into therapeutics. It provides an in-depth exploration of the evolving landscape of therapeutic development, which is currently advancing from bench to bedside. These ongoing efforts hold the promise of significantly transforming the lives of vestibular schwannoma patients in the future.
Collapse
Affiliation(s)
- Shaolei Guo
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuan Zheng
- Department of Neurosurgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenli Chen
- Department of Neurosurgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Umar Raza
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, China
| | - Ailiang Zeng
- Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, Texas, USA
| | - Farhana Akter
- Faculty of Arts and Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Quan Huang
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shun Yao
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Advances in Targeted Therapy for Neurofibromatosis Type 2 (NF2)-Associated Vestibular Schwannomas. Curr Oncol Rep 2023; 25:531-537. [PMID: 36933171 DOI: 10.1007/s11912-023-01388-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 03/19/2023]
Abstract
PURPOSE OF REVIEW Neurofibromatosis 2 (NF2) is an autosomal-dominant genetic disorder characterized by bilateral vestibular schwannomas (VS), meningiomas, ependymomas, spinal and peripheral schwannomas, optic gliomas, and juvenile cataracts. Ongoing studies provide new insight into the role of the NF2 gene and merlin in VS tumorigenesis. RECENT FINDINGS As NF2 tumor biology becomes increasingly understood, therapeutics targeting specific molecular pathways have been developed and evaluated in preclinical and clinical studies. NF2-associated VS are a source of significant morbidity with current treatments including surgery, radiation, and observation. Currently, there are no FDA-approved medical therapies for VS, and the development of selective therapeutics is a high priority. This manuscript reviews NF2 tumor biology and current therapeutics undergoing investigation for treatment of patients with VS.
Collapse
|
3
|
Dasatinib enhances curcumin-induced cytotoxicity, apoptosis and protective autophagy in human schwannoma cells HEI-193: The role of Akt/mTOR/p70S6K signalling pathway. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2022; 72:403-414. [PMID: 36651538 DOI: 10.2478/acph-2022-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 01/26/2023]
Abstract
The present study was carried out in human schwannoma cells (HEI-193) to determine the combined anti-cancer effect of curcumin and dasatinib. Cells were treated with curcumin only, dasatinib only, or the combination of curcumin and dasatinib for 24 hours. Cellular toxicity, cell proliferation, and cell death were determined by LDH, MTT, and trypan blue dye assays, respectively. ELISA based kit was used to determine apoptotic cell death. Western blotting was used to determine the expression of apoptotic and autophagy-associated protein markers. Similarly, expression levels of Akt/mTOR/p70S6K signalling pathway-related proteins were studied using Western blotting. Cell death and apoptosis were significantly higher in HEI-193 cells treated with curcumin and dasatinib combination compared to individual controls. The combination of curcumin and dasatinib significantly enhances autophagy markers compared to individual controls. Furthermore, the combination of curcumin and dasatinib significantly activates Akt/mTOR/p70S6K signalling pathway compared to individual controls. In conclusion, our results suggest that the combination of curcumin and dasatinib significantly enhances cytotoxicity, apoptosis, and protective autophagy in HEI-193 cells through Akt/mTOR/p70S6K signalling pathway.
Collapse
|
4
|
Alghriany AI, Omar HELDM, Mahmoud AM, Atia MM. Assessment of the Toxicity of Aluminum Oxide and Its Nanoparticles in the Bone Marrow and Liver of Male Mice: Ameliorative Efficacy of Curcumin Nanoparticles. ACS OMEGA 2022; 7:13841-13852. [PMID: 35559158 PMCID: PMC9088947 DOI: 10.1021/acsomega.2c00195] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/07/2022] [Indexed: 05/24/2023]
Abstract
The potential influence of nanoparticles (NPs) on the liver and bone marrow has received attention. The aim of this work was to evaluate the effect of nanocurcumin on the oxidative stress, apoptosis, and toxicity induced by Al2O3 and its NPs. The experimental animals (n = 72 mice) were divided into the following groups: group I, as a control; groups II and III, as aluminum oxide and its NPs (6 mg/kg); group IV, as aluminum oxide + nanocurcumin (Al2O3 + N-Cur, 20 mg/kg); and group V, as aluminum oxide NPs + nanocurcumin (Al2O3-NP + N.Cur., 20 mg/kg). Al2O3 and its NP groups significantly increased p53, Nrf2 levels, and the white blood cell count. They also decreased the Hsp70 level, antitrypsin, immunoglobulin G, and the red blood cell count. In addition, they significantly decreased the total and differential bone marrow cell counts and the maturation index ratio (MIR). Nanocurcumin (N.Cur.) reverted the previous proteins, blood parameters, total bone marrow cell count, and the MIR as M/E, I/Mg, MMI, I/Me, and EMI to normal. Furthermore, N.Cur. prevented apoptosis and reduced the histopathological score and collagen fiber percentage caused by Al2O3 and its NPs in the liver. Nanotechnology was used to increase the therapeutic efficiency of curcumin against the harmful effects of oxidative stress associated with Al2O3 NPs.
Collapse
Affiliation(s)
- Alshaimaa
A. I. Alghriany
- Laboratory
of Molecular Cell Biology; Department of Zoology, Faculty of Science, Assiut University, Assiut 71515, Egypt
| | - Hossam EL-din M. Omar
- Laboratory
of Physiology, Department of Zoology, Faculty of Science, Assiut University, Assiut 71515, Egypt
| | - Amera M. Mahmoud
- Laboratory
of Physiology, Department of Zoology, Faculty of Science, Assiut University, Assiut 71515, Egypt
| | - Mona M. Atia
- Laboratory
of Molecular Cell Biology; Department of Zoology, Faculty of Science, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
5
|
Amaravathi A, Oblinger JL, Welling DB, Kinghorn AD, Chang LS. Neurofibromatosis: Molecular Pathogenesis and Natural Compounds as Potential Treatments. Front Oncol 2021; 11:698192. [PMID: 34604034 PMCID: PMC8485038 DOI: 10.3389/fonc.2021.698192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022] Open
Abstract
The neurofibromatosis syndromes, including NF1, NF2, and schwannomatosis, are tumor suppressor syndromes characterized by multiple nervous system tumors, particularly Schwann cell neoplasms. NF-related tumors are mainly treated by surgery, and some of them have been treated by but are refractory to conventional chemotherapy. Recent advances in molecular genetics and genomics alongside the development of multiple animal models have provided a better understanding of NF tumor biology and facilitated target identification and therapeutic evaluation. Many targeted therapies have been evaluated in preclinical models and patients with limited success. One major advance is the FDA approval of the MEK inhibitor selumetinib for the treatment of NF1-associated plexiform neurofibroma. Due to their anti-neoplastic, antioxidant, and anti-inflammatory properties, selected natural compounds could be useful as a primary therapy or as an adjuvant therapy prior to or following surgery and/or radiation for patients with tumor predisposition syndromes, as patients often take them as dietary supplements and for health enhancement purposes. Here we review the natural compounds that have been evaluated in NF models. Some have demonstrated potent anti-tumor effects and may become viable treatments in the future.
Collapse
Affiliation(s)
- Anusha Amaravathi
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Janet L Oblinger
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - D Bradley Welling
- Department of Otolaryngology Head & Neck Surgery, Harvard Medical School, Massachusetts Eye and Ear, and Massachusetts General Hospital, Boston, MA, United States
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University College of Pharmacy, Columbus, OH, United States
| | - Long-Sheng Chang
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States.,Department of Otolaryngology-Head & Neck Surgery, The Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
6
|
Zoltán O, László P, Éva K, Béla V. A "keto-enol" plaque buster mechanism to diminish Alzheimer's β-Amyloid burden. Biochem Biophys Res Commun 2020; 532:82-87. [PMID: 32828536 DOI: 10.1016/j.bbrc.2020.07.086] [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: 06/26/2020] [Accepted: 07/17/2020] [Indexed: 11/26/2022]
Abstract
Curcumin and related compounds have been validated to remove even well-developed human β-amyloid plaques from the brain of transgenic mice, in vivo. However, their molecular mechanism of the plaque buster activity is rather unknown. Computational chemistry was employed here to better understand the β-amyloid protein elimination. According to our docking studies, a tautomeric "keto-enol" flip-flop mechanism is proposed that may chop up β-amyloid plaques in Alzheimer's due to removing each hairpin-foldamers one by one from both ends of aggregated fibrils. According to the experimented models, other bi-stable "keto-enol" pharmacophores might be identified to break up amyloid plaques and enhance rapid clearance of toxic aggregates in Alzheimer's disease.
Collapse
Affiliation(s)
- Oláh Zoltán
- Bioinformatics and Molecular Surgery Unit, Institute of Chemistry, Faculty of Materials Science and Engineering, University of Miskolc, Miskolc, Hungary; Acheuron Ltd, Szeged, Hungary; Forget-Me-Not B2B Ltd, Szeged, Hungary
| | | | - Kocsis Éva
- Bioinformatics and Molecular Surgery Unit, Institute of Chemistry, Faculty of Materials Science and Engineering, University of Miskolc, Miskolc, Hungary; Acheuron Ltd, Szeged, Hungary; Forget-Me-Not B2B Ltd, Szeged, Hungary
| | - Viskolcz Béla
- Bioinformatics and Molecular Surgery Unit, Institute of Chemistry, Faculty of Materials Science and Engineering, University of Miskolc, Miskolc, Hungary
| |
Collapse
|
7
|
Perrone L, Sampaolo S, Melone MAB. Bioactive Phenolic Compounds in the Modulation of Central and Peripheral Nervous System Cancers: Facts and Misdeeds. Cancers (Basel) 2020; 12:cancers12020454. [PMID: 32075265 PMCID: PMC7072310 DOI: 10.3390/cancers12020454] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
Abstract
Efficacious therapies are not available for the cure of both gliomas and glioneuronal tumors, which represent the most numerous and heterogeneous primary cancers of the central nervous system (CNS), and for neoplasms of the peripheral nervous system (PNS), which can be divided into benign tumors, mainly represented by schwannomas and neurofibromas, and malignant tumors of the peripheral nerve sheath (MPNST). Increased cellular oxidative stress and other metabolic aspects have been reported as potential etiologies in the nervous system tumors. Thus polyphenols have been tested as effective natural compounds likely useful for the prevention and therapy of this group of neoplasms, because of their antioxidant and anti-inflammatory activity. However, polyphenols show poor intestinal absorption due to individual intestinal microbiota content, poor bioavailability, and difficulty in passing the blood-brain barrier (BBB). Recently, polymeric nanoparticle-based polyphenol delivery improved their gastrointestinal absorption, their bioavailability, and entry into defined target organs. Herein, we summarize recent findings about the primary polyphenols employed for nervous system tumor prevention and treatment. We describe the limitations of their application in clinical practice and the new strategies aimed at enhancing their bioavailability and targeted delivery.
Collapse
Affiliation(s)
- Lorena Perrone
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, Via Sergio Pansini, 5 80131 Naples, Italy; (L.P.); (S.S.)
- Department of Chemistry and Biology, University Grenoble Alpes, 38400 Saint-Martin-d’Hères, France
| | - Simone Sampaolo
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, Via Sergio Pansini, 5 80131 Naples, Italy; (L.P.); (S.S.)
| | - Mariarosa Anna Beatrice Melone
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, Via Sergio Pansini, 5 80131 Naples, Italy; (L.P.); (S.S.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Temple University, BioLife Building (015-00)1900 North 12th Street, Philadelphia, PA 19122-6078, USA
- Correspondence:
| |
Collapse
|
8
|
Liposome Delivery of Natural STAT3 Inhibitors for the Treatment of Cancer. PHARMACEUTICAL FRONTIERS 2019; 1. [PMID: 31886474 DOI: 10.20900/pf20190007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In the tumor microenvironment, cytokines, growth factors, and oncogenes mediate constitutive activation of the signal transducer and activator of transcription 3 (STAT3) signaling pathway in both cancer cells and infiltrating immune cells. STAT3 activation in cancer cells drives tumorigenic changes that allow for increased survival, proliferation, and resistance to apoptosis. The modulation of immune cells is more complicated and conflicting. STAT3 signaling drives the myeloid cell phenotype towards an immune suppressive state, which mediates T cell inhibition. On the other hand, STAT3 signaling in T cells leads to proliferation and T cell activity required for an anti-tumor response. Targeted delivery of STAT3 inhibitors to cancer cells and myeloid cells could therefore improve therapeutic outcomes. Many compounds that inhibit the STAT3 pathways for cancer treatment include peptide drugs, small molecule inhibitors, and natural compounds. However, natural compounds that inhibit STAT3 are often hydrophobic, which reduces their bioavailability and leads to unfavorable pharmacokinetics. This review focuses specifically on liposome-encapsulated natural STAT3 inhibitors and their ability to target cancer cells and myeloid cells to reduce tumor growth and decrease STAT3-mediated immune suppression. Many of these liposome formulations have led to profound tumor reduction and examples of combination formulations have been shown to eliminate tumors through immune modulation.
Collapse
|
9
|
Forouzanfar F, Barreto G, Majeed M, Sahebkar A. Modulatory effects of curcumin on heat shock proteins in cancer: A promising therapeutic approach. Biofactors 2019; 45:631-640. [PMID: 31136038 DOI: 10.1002/biof.1522] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/12/2019] [Indexed: 12/23/2022]
Abstract
Cancer metastasis represents a multistep process, including alteration of cell adhesion/motility in the microenvironment and sustained angiogenesis, which is essential for supporting cancer growth in tissues that are distant from the primary tumor. There is growing evidence suggesting that heat shock proteins (HSPs) (also known as heat stress proteins), which constitute a family of stress-inducible proteins, may be involved in the pathogenesis of cancer. Curcumin (diferuloylmethane) is a potent anti-inflammatory, antioxidant, antimicrobial, and antitumor agent. Curcumin has been shown to regulate different members of HSPs including HSP27, HSP40, HSP60, HSP70, and HSP90 in cancer. Here, we present extent findings suggesting that curcumin may act as a potential therapeutic agent for the treatment of cancer through its regulation of HSPs.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - George Barreto
- Departamento de Nutrición yBioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | | | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology,School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Sohn EJ, Bak KM, Nam YK, Park HT. Upregulation of microRNA 344a-3p is involved in curcumin induced apoptosis in RT4 schwannoma cells. Cancer Cell Int 2018; 18:199. [PMID: 30534000 PMCID: PMC6278133 DOI: 10.1186/s12935-018-0693-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 11/27/2018] [Indexed: 12/14/2022] Open
Abstract
Background Schwannoma arising from peripheral nervous sheaths is a benign tumor. Methods To evaluate cell cytotoxicity, (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) tetrazolium reduction and terminal deoxynucleotidyltransferase UTP nick-end labeling (TUNEL) assays were used. A microRNA (miRNA) array was used to identify the miRNAs involved in curcumin-induced apoptosis. To examine miRNA expression, quantitative RT-PCR was used. Results In this study, curcumin exerted cellular cytotoxicity against RT4 schwannoma cells, with an increase in TUNEL-positive cells. Curcumin also activated the expression of apoptotic proteins, such as polyADP ribose polymerase, caspase-3, and caspase-9. The miRNA array revealed that seven miRNAs (miRNA 350, miRNA 17-2-3p, let 7e-3p, miRNA1224, miRNA 466b-1-3p, miRNA 18a-5p, and miRNA 322-5p) were downregulated following treatment with both 10 and 20 μM curcumin in RT4 cells, while four miRNAs (miRNA122-5p, miRNA 3473, miRNA182, and miRNA344a-3p) were upregulated. Interestingly, transfection with a miRNA 344a-3p mimic downregulated the mRNA expression of Bcl2 and upregulated that of Bax, Curcumin treatment in RT 4 cells also reduced the mRNA expression of Bcl2 and enhanced expression of Bax, Overexpression of miRNA344a-3p mimic combined with curcumin treatment activated the expression of apoptotic proteins, including procaspase-9 and cleaved caspase-3 while inhibition of miRNA 344a-3p using miR344a-3p inhibitor repressed cleaved caspase-3 and -9 in curcumin treated RT-4 cells compared to control. Conclusions Our findings demonstrate that curcumin induces apoptosis in schwannoma cells via miRNA 344a-3p. Thus, curcumin may serve as a potent therapeutic agent for the treatment of schwannoma.
Collapse
Affiliation(s)
- Eun Jung Sohn
- Peripheral Neuropathy Research Center, Department of Molecular Neuroscience, College of Medicine, Dong-A University, Dongdaesin-Dong, Seo-Gu, Busan, 602-714 South Korea
| | - Kyoung-Mi Bak
- Peripheral Neuropathy Research Center, Department of Molecular Neuroscience, College of Medicine, Dong-A University, Dongdaesin-Dong, Seo-Gu, Busan, 602-714 South Korea
| | - Yun-Kyeong Nam
- Peripheral Neuropathy Research Center, Department of Molecular Neuroscience, College of Medicine, Dong-A University, Dongdaesin-Dong, Seo-Gu, Busan, 602-714 South Korea
| | - Hwan Tae Park
- Peripheral Neuropathy Research Center, Department of Molecular Neuroscience, College of Medicine, Dong-A University, Dongdaesin-Dong, Seo-Gu, Busan, 602-714 South Korea
| |
Collapse
|
11
|
Kiss A, Erdődi F, Lontay B. Myosin phosphatase: Unexpected functions of a long-known enzyme. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:2-15. [PMID: 30076859 DOI: 10.1016/j.bbamcr.2018.07.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/09/2018] [Accepted: 07/26/2018] [Indexed: 01/08/2023]
Abstract
Myosin phosphatase (MP) holoenzyme is a Ser/Thr specific enzyme, which is the member of protein phosphatase type 1 (PP1) family and composed of a PP1 catalytic subunit (PP1c/PPP1CB) and a myosin phosphatase targeting subunit (MYPT1/PPP1R12A). PP1c is required for the catalytic activity of the holoenzyme, while MYPT1 regulates MP through targeting the holoenzyme to its substrates. Above the well-characterized function of MP, as the major regulator of smooth muscle contractility mediating the dephosphorylation of 20 kDa myosin light chain, accumulating data support its role in other, non-contractile functions. In this review, we summarize the scaffold function of MP holoenzyme and its roles in processes such as cell cycle, development, gene expression regulation and neurotransmitter release. In particular, we highlight novel interacting proteins of MYPT1 and pathophysiological functions of MP relevant to tumorigenesis, insulin resistance and neurodegenerative disorders. This article is part of a Special Issue entitled: Protein Phosphatases as Critical Regulators for Cellular Homeostasis edited by Prof. Peter Ruvolo and Dr. Veerle Janssens.
Collapse
Affiliation(s)
- Andrea Kiss
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ferenc Erdődi
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Beáta Lontay
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
12
|
Hu Y, Fu A, Miao Z, Zhang X, Wang T, Kang A, Shan J, Zhu D, Li W. Fluorescent ligand fishing combination with in-situ imaging and characterizing to screen Hsp 90 inhibitors from Curcuma longa L. based on InP/ZnS quantum dots embedded mesoporous nanoparticles. Talanta 2018; 178:258-267. [DOI: 10.1016/j.talanta.2017.09.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/05/2017] [Accepted: 09/12/2017] [Indexed: 01/08/2023]
|
13
|
Squillaro T, Schettino C, Sampaolo S, Galderisi U, Di Iorio G, Giordano A, Melone MAB. Adult‐onset brain tumors and neurodegeneration: Are polyphenols protective? J Cell Physiol 2017; 233:3955-3967. [DOI: 10.1002/jcp.26170] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Tiziana Squillaro
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in NeurosciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Carla Schettino
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in NeurosciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Simone Sampaolo
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in NeurosciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology and Molecular Biology SectionUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Giuseppe Di Iorio
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in NeurosciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Antonio Giordano
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and TechnologyTemple UniversityPhiladelphiaPennsylvania
- Department of MedicineSurgery and Neuroscience University of SienaSienaItaly
| | - Mariarosa A. B. Melone
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in NeurosciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and TechnologyTemple UniversityPhiladelphiaPennsylvania
| |
Collapse
|
14
|
Sahin K, Pala R, Tuzcu M, Ozdemir O, Orhan C, Sahin N, Juturu V. Curcumin prevents muscle damage by regulating NF-κB and Nrf2 pathways and improves performance: an in vivo model. J Inflamm Res 2016; 9:147-54. [PMID: 27621662 PMCID: PMC5010171 DOI: 10.2147/jir.s110873] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose Exercise (Ex) increases reactive oxygen species and impairs antioxidant defense systems. Recent data suggest that curcumin (CW) possesses peroxisome proliferator-activated receptor gamma activity and anti-inflammatory properties. Therefore, this study was designed to investigate the effects of CW supplementation on Ex performance, endurance, and changes in serum and muscle proteins in rats after exhaustive Ex. Materials and methods Twenty-eight (28) male Wistar rats (age: 8 weeks and body weight: 180±20 g) were divided into four treatment groups: 1) control (C; no Ex), 2) C + CW (no Ex + CW), 3) C + Ex, and 4) C + Ex + CW (Ex + CW). CW was administered as 100 mg/kg CurcuWin®, providing 20 mg of curcuminoids daily for 6 weeks. A motor-driven rodent treadmill was used to carry out the Ex protocols. During a 5-day period, animals in chronic Ex groups were put through different regimens: day 1, 10 m/min for 10 minutes; day 2, 20 m/min for 10 minutes; day 3, 25 m/min for 10 minutes; day 4, 25 m/min for 20 minutes; and day 5, 25 m/min for 30 minutes. Animals were exercised at 25 m/min for 45 min/d for 5 d/wk for 6 weeks. Blood and muscle samples were analyzed for muscle markers, oxidative stress, and antioxidant markers. Results Lactate and muscle malondialdehyde levels decreased in the CW-treated groups (P<0.0001). However, activities of antioxidant enzyme levels increased in the CW-treated groups. Run to exhaustion (minutes) improved in the CW-treated groups. Muscle nuclear factor-κB (P<0.05) and heat shock protein 70 (P<0.05) levels were much lowered in the CW treated group followed by Ex group. In addition, muscle inhibitors of kappa B, peroxisome proliferator-activated receptor gamma coactivator 1-alpha, thioredoxin-1, sirtuin 1, nuclear factor (erythroid-derived 2)-like 2, and glucose transporter 4 protein levels in the Ex + CW group were higher than those in the control and Ex groups (P<0.05). Conclusion This study suggests that novel CW has the potential to help prevent muscle damage by regulating the nuclear factor-κB and nuclear factor (erythroid-derived 2)-like 2 pathways and improve the performance and nutritional values of CW.
Collapse
Affiliation(s)
- Kazim Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine
| | - Ragip Pala
- Department of Movement and Training Science
| | - Mehmet Tuzcu
- Department of Biology, Firat University, Elazig, Turkey
| | | | - Cemal Orhan
- Department of Animal Nutrition, Faculty of Veterinary Medicine
| | - Nurhan Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine
| | - Vijaya Juturu
- OmniActive Health Technologies Inc., Morristown, NJ, USA
| |
Collapse
|
15
|
Radons J. The human HSP70 family of chaperones: where do we stand? Cell Stress Chaperones 2016; 21:379-404. [PMID: 26865365 PMCID: PMC4837186 DOI: 10.1007/s12192-016-0676-6] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 01/23/2023] Open
Abstract
The 70-kDa heat shock protein (HSP70) family of molecular chaperones represents one of the most ubiquitous classes of chaperones and is highly conserved in all organisms. Members of the HSP70 family control all aspects of cellular proteostasis such as nascent protein chain folding, protein import into organelles, recovering of proteins from aggregation, and assembly of multi-protein complexes. These chaperones augment organismal survival and longevity in the face of proteotoxic stress by enhancing cell viability and facilitating protein damage repair. Extracellular HSP70s have a number of cytoprotective and immunomodulatory functions, the latter either in the context of facilitating the cross-presentation of immunogenic peptides via major histocompatibility complex (MHC) antigens or in the context of acting as "chaperokines" or stimulators of innate immune responses. Studies have linked the expression of HSP70s to several types of carcinoma, with Hsp70 expression being associated with therapeutic resistance, metastasis, and poor clinical outcome. In malignantly transformed cells, HSP70s protect cells from the proteotoxic stress associated with abnormally rapid proliferation, suppress cellular senescence, and confer resistance to stress-induced apoptosis including protection against cytostatic drugs and radiation therapy. All of the cellular activities of HSP70s depend on their adenosine-5'-triphosphate (ATP)-regulated ability to interact with exposed hydrophobic surfaces of proteins. ATP hydrolysis and adenosine diphosphate (ADP)/ATP exchange are key events for substrate binding and Hsp70 release during folding of nascent polypeptides. Several proteins that bind to distinct subdomains of Hsp70 and consequently modulate the activity of the chaperone have been identified as HSP70 co-chaperones. This review focuses on the regulation, function, and relevance of the molecular Hsp70 chaperone machinery to disease and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jürgen Radons
- Scientific Consulting International, Mühldorfer Str. 64, 84503, Altötting, Germany.
| |
Collapse
|
16
|
Sun L, Wan K, Hu X, Zhang Y, Yan Z, Feng J, Zhang J. Functional nanoemulsion-hybrid lipid nanocarriers enhance the bioavailability and anti-cancer activity of lipophilic diferuloylmethane. NANOTECHNOLOGY 2016; 27:085102. [PMID: 26808001 DOI: 10.1088/0957-4484/27/8/085102] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The purpose of this study was to assess the enhanced physicochemical characteristics, in vitro release behavior, anti-lung cancer activity, gastrointestinal absorption, in vivo bioavailability and bioequivalence of functional nanoemulsion-hybrid lipid nanocarriers containing diferuloylmethane (DNHLNs). The DNHLNs were first fabricated by loading water-in-oil nanoemulsions into hybrid lipid nanosystems using nanoemulsion-thin film-sonication dispersion technologies. The in situ absorption and in vitro and in vivo kinetic features of DNHLNs were measured using an in situ unidirectional perfusion method, a dynamic dialysis method and a plasma concentration-time profile-based method, respectively. The cytotoxic effects of DNHLNs in lung adenocarcinoma A549 cells were examined using MTT colorimetric analysis. The absorptive constants and permeabilities of DNHLNs in four gastrointestinal sections increased by 1.43-3.23 times and by 3.10-7.76 times that of diferuloylmethane (DIF), respectively. The relative bioavailability of DNHLNs to free DIF was 855.02%. DNHLNs inhibited cancer cell growth in a time- and dose-dependent manner. DNHLNs markedly improved the absorption and bioavailability of DIF after oral administration. DNHLNs had stronger inhibitory effects on the viability of A549 cells than that of free DIF. DNHLNs might be potentially promising nanocarriers for DIF delivery via the oral route to address unmet clinical needs.
Collapse
|
17
|
Ashraf A, Pervaiz S. Hippo circuitry and the redox modulation of hippo components in cancer cell fate decisions. Int J Biochem Cell Biol 2015; 69:20-8. [PMID: 26456518 DOI: 10.1016/j.biocel.2015.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 10/02/2015] [Accepted: 10/03/2015] [Indexed: 12/17/2022]
Abstract
Meticulous and precise control of organ size is undoubtedly one of the most pivotal processes in mammalian development and regeneration along with cell differentiation, morphogenesis and programmed cell death. These processes are strictly regulated by complex and highly coordinated mechanisms to maintain a steady growth state. There are a number of extrinsic and intrinsic factors that dictate the total number and/or size of cells by influencing growth, proliferation, differentiation and cell death. Multiple pathways, such as those involved in promoting organ size and others that restrict disproportionate tissue growth act simultaneously to maintain cellular and tissue homeostasis. Aberrations at any level in these organ size-regulating processes can lead to various pathological states with cancers being the most formidable one (Yin and Zhang, 2011). Extensive research in the realm of growth control has led to the identification of the Hippo-signaling pathway as a critical network in modulating tissue growth via its effect on multiple signaling pathways and through intricate crosstalk with proteins that regulate cell polarity, adhesion and cell-cell interactions (Zhao et al., 2011b). The Hippo pathway controls cell number and organ size by transducing signals from the plasma membrane to the nucleus to regulate the expression of genes involved in cell fate determination (Shi et al., 2015). In this review, we summarize the recent discoveries concerning Hippo pathway, its diversiform regulation in mammals as well as its implications in cancers, and highlight the possible role of oxidative stress in Hippo pathway regulation.
Collapse
Affiliation(s)
- Asma Ashraf
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; School of Biomedical Sciences, Curtin University, Perth, Australia.
| |
Collapse
|
18
|
Dilwali S, Briët MC, Kao SY, Fujita T, Landegger LD, Platt MP, Stankovic KM. Preclinical validation of anti-nuclear factor-kappa B therapy to inhibit human vestibular schwannoma growth. Mol Oncol 2015; 9:1359-70. [PMID: 25891780 DOI: 10.1016/j.molonc.2015.03.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 02/22/2015] [Accepted: 03/23/2015] [Indexed: 01/25/2023] Open
Abstract
Vestibular schwannomas (VSs), the most common tumors of the cerebellopontine angle, arise from Schwann cells lining the vestibular nerve. Pharmacotherapies against VS are almost non-existent. Although the therapeutic inhibition of inflammatory modulators has been established for other neoplasms, it has not been explored in VS. A bioinformatic network analysis of all genes reported to be differentially expressed in human VS revealed a pro-inflammatory transcription factor nuclear factor-kappa B (NF-κB) as a central molecule in VS pathobiology. Assessed at the transcriptional and translational level, canonical NF-κB complex was aberrantly activated in human VS and derived VS cultures in comparison to control nerves and Schwann cells, respectively. Cultured primary VS cells and VS-derived human cell line HEI-193 were treated with specific NF-κB siRNAs, experimental NF-κB inhibitor BAY11-7082 (BAY11) and clinically relevant NF-κB inhibitor curcumin. Healthy human control Schwann cells from the great auricular nerve were also treated with BAY11 and curcumin to assess toxicity. All three treatments significantly reduced proliferation in primary VS cultures and HEI-193 cells, with siRNA, 5 μM BAY11 and 50 μM curcumin reducing average proliferation (±standard error of mean) to 62.33% ± 10.59%, 14.3 ± 9.7%, and 23.0 ± 20.9% of control primary VS cells, respectively. These treatments also induced substantial cell death. Curcumin, unlike BAY11, also affected primary Schwann cells. This work highlights NF-κB as a key modulator in VS cell proliferation and survival and demonstrates therapeutic efficacy of directly targeting NF-κB in VS.
Collapse
Affiliation(s)
- Sonam Dilwali
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Harvard-MIT Program in Speech and Hearing Bioscience and Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Martijn C Briët
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Otorhinolaryngology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Shyan-Yuan Kao
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA.
| | - Takeshi Fujita
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Otology and Laryngology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| | - Lukas D Landegger
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Otology and Laryngology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| | - Michael P Platt
- Department of Otology and Laryngology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Department of Otolaryngology-Head and Neck Surgery, Boston University, 72 E Concord Street, Boston, MA 02118, USA.
| | - Konstantina M Stankovic
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Harvard-MIT Program in Speech and Hearing Bioscience and Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Otology and Laryngology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| |
Collapse
|
19
|
Schroeder RD, Angelo LS, Kurzrock R. NF2/merlin in hereditary neurofibromatosis 2 versus cancer: biologic mechanisms and clinical associations. Oncotarget 2014; 5:67-77. [PMID: 24393766 PMCID: PMC3960189 DOI: 10.18632/oncotarget.1557] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Inactivating germline mutations in the tumor suppressor gene NF2 cause the hereditary syndrome neurofibromatosis 2, which is characterized by the development of neoplasms of the nervous system, most notably bilateral vestibular schwannoma. Somatic NF2 mutations have also been reported in a variety of cancers, but interestingly these mutations do not cause the same tumors that are common in hereditary neurofibromatosis 2, even though the same gene is involved and there is overlap in the site of mutations. This review highlights cancers in which somatic NF2 mutations have been found, the cell signaling pathways involving NF2/merlin, current clinical trials treating neurofibromatosis 2 patients, and preclinical findings that promise to lead to new targeted therapies for both cancers harboring NF2 mutations and neurofibromatosis 2 patients.
Collapse
Affiliation(s)
- Rebecca Dunbar Schroeder
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | |
Collapse
|
20
|
Banerjee S, Sahoo AK, Chattopadhyay A, Ghosh SS. Recombinant IκBα-loaded curcumin nanoparticles for improved cancer therapeutics. NANOTECHNOLOGY 2014; 25:345102. [PMID: 25091588 DOI: 10.1088/0957-4484/25/34/345102] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The field of recombinant protein therapeutics has been evolving rapidly, making significant impact on clinical applications for several diseases, including cancer. However, the functional aspects of proteins rely exclusively on their structural integrity, in which nanoparticle mediated delivery offers unique advantages over free proteins. In the present work, a novel strategy has been developed where the nanoparticles (NPs) used for the delivery of the recombinant protein could contribute to enhancing the therapeutic efficacy of the recombinant protein. The transcription factor, NFκB, involved in cell growth and its inhibitor, IκBα, regulates its proliferation. Another similar naturally available molecule, which inhibits the function of NFκB, is curcumin. Hence, we have developed a 'green synthesis' method for preparing water-soluble curcumin nanoparticles to stabilize recombinant IκBα protein. The NPs were characterized by UV-vis and fluorescence spectroscopy, transmission electron microscopy (TEM) and dynamic light scattering before administration into human cervical carcinoma (HeLa) and glioblastoma (U87MG) cells. Experimental results demonstrated that this combined module had enhanced therapeutic efficacy, causing apoptotic cell death, which was confirmed by cytotoxicity assay and flowcytometry analyses. The expression of apoptotic genes studied by semi-quantitative reverse transcription PCR delineated the molecular pathways involved in cell death. Thus, our study revealed that the functional delivery of recombinant IκBα-loaded curcumin NPs has promise as a natural-product-based protein therapeutics against cancer cells.
Collapse
Affiliation(s)
- Subhamoy Banerjee
- Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | | | | | | |
Collapse
|
21
|
Lim SHS, Ardern-Holmes S, McCowage G, de Souza P. Systemic therapy in neurofibromatosis type 2. Cancer Treat Rev 2014; 40:857-61. [PMID: 24877986 DOI: 10.1016/j.ctrv.2014.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 01/06/2023]
Abstract
The systemic treatment of patients with neurofibromatosis type 2 associated tumours is challenging, as these patients often have prolonged survival but with the inevitable propensity for their disease to cause symptoms, and no effective therapies other than local treatments such as surgery. Understanding the molecular mechanisms driving NF-2 pathogenesis holds promise for the potential use of targeted therapy. Initial studies of agents such as bevacizumab (angiogenesis inhibitor) and lapatinib (epidermal growth factor and ErbB2 inhibitor) have indicated benefit for selected patients. As the biology of NF-2 is dependent on multiple interlinked downstream signalling pathways, targeting multiple pathways may be more effective than single agents. Phase zero trials, adaptive phase II or small multi-arm trials, are likely the way forward in this rare disease. Ideally, well-tolerated targeted therapy would appear to be the most promising approach for patients with NF-2, given the natural history of this disease.
Collapse
Affiliation(s)
- Stephanie Hui-Su Lim
- Department of Medical Oncology and Ingham Research Institute, Liverpool, NSW, Australia.
| | - Simone Ardern-Holmes
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, Westmead, NSW, Australia.
| | - Geoffrey McCowage
- Oncology Research Unit, The Children's Hospital at Westmead, Westmead, NSW, Australia.
| | - Paul de Souza
- Department of Medical Oncology and Ingham Research Institute, Liverpool, NSW, Australia; University of Western Sydney, School of Medicine, Molecular Medicine Research Group, NSW, Australia.
| |
Collapse
|
22
|
Natural compounds as potential treatments of NF2-deficient schwannoma and meningioma: cucurbitacin D and goyazensolide. Otol Neurotol 2014; 34:1519-27. [PMID: 23928514 DOI: 10.1097/mao.0b013e3182956169] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
HYPOTHESIS Cucurbitacin D and goyazensolide, 2 plant-derived natural compounds, possess potent growth-inhibitory activity in schwannoma and meningioma cells. BACKGROUND Currently, no FDA-approved drugs are available for neurofibromatosis type 2 (NF2)-associated schwannomas and meningiomas. Selected natural compounds with antineoplastic activity, such as cucurbitacin D and goyazensolide, may be developed as potential treatments for these tumors. METHODS The Nf2-deficient mouse schwannoma Sch10545 and human benign meningioma Ben-Men-1 cells were treated with various concentrations of cucurbitacin D and goyazensolide. The effect on cell proliferation was determined using resazurin assays. Flow cytometry was used to assess the cell cycle profiles. Western blot analysis was performed to investigate the expression of various signaling molecules related to the cell cycle and the AKT pathway. RESULTS Cucurbitacin D inhibited proliferation of Sch10545 cells (IC50 ∼ 0.75 μM) and Ben-Men-1 cells (IC50 ∼0.2 μM). Goyazensolide also reduced cell proliferation of Sch10545 cells (IC50 ∼0.9 μM) and Ben-Men-1 cells (IC50 ∼1 μM). The G2/M population increased in both Sch10545 and Ben-Men-1 cells treated with cucurbitacin D or goyazensolide around the IC50. Cucurbitacin and goyazensolide substantially reduced the levels of cyclins E and A in treated Sch10545 and Ben-Men-1 cells. Cucurbitacin D also inhibited cyclin B, phospho-AKT and phospho-PRAS40 expression. In addition, goyazensolide reduced the levels of phospho-AKT and NFκB and increased the expression of pro-apoptotic Bim in Sch10545 and Ben-Men-1 cells. CONCLUSION Both cucurbitacin D and goyazensolide effectively inhibit proliferation of NF2-deficient schwannoma and meningioma cells, suggesting that these natural compounds should be further evaluated as potential treatments for NF2-related tumors.
Collapse
|
23
|
Firouzi Z, Lari P, Rashedinia M, Ramezani M, Iranshahi M, Abnous K. Proteomics screening of molecular targets of curcumin in mouse brain. Life Sci 2014; 98:12-7. [DOI: 10.1016/j.lfs.2013.12.200] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/22/2013] [Accepted: 12/23/2013] [Indexed: 01/31/2023]
|
24
|
|
25
|
Tanaka K, Eskin A, Chareyre F, Jessen WJ, Manent J, Niwa-Kawakita M, Chen R, White CH, Vitte J, Jaffer ZM, Nelson SF, Rubenstein AE, Giovannini M. Therapeutic potential of HSP90 inhibition for neurofibromatosis type 2. Clin Cancer Res 2013; 19:3856-70. [PMID: 23714726 DOI: 10.1158/1078-0432.ccr-12-3167] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The growth and survival of neurofibromatosis type 2 (NF2)-deficient cells are enhanced by the activation of multiple signaling pathways including ErbBs/IGF-1R/Met, PI3K/Akt, and Ras/Raf/Mek/Erk1/2. The chaperone protein HSP90 is essential for the stabilization of these signaling molecules. The aim of the study was to characterize the effect of HSP90 inhibition in various NF2-deficient models. EXPERIMENTAL DESIGN We tested efficacy of the small-molecule NXD30001, which has been shown to be a potent HSP90 inhibitor. The antiproliferative activity of NXD30001 was tested in NF2-deficient cell lines and in human primary schwannoma and meningioma cultures in vitro. The antitumor efficacy of HSP90 inhibition in vivo was verified in two allograft models and in one NF2 transgenic model. The underlying molecular alteration was further characterized by a global transcriptome approach. RESULTS NXD30001 induced degradation of client proteins in and suppressed proliferation of NF2-deficient cells. Differential expression analysis identified subsets of genes implicated in cell proliferation, cell survival, vascularization, and Schwann cell differentiation whose expression was altered by NXD30001 treatment. The results showed that NXD30001 in NF2-deficient schwannoma suppressed multiple pathways necessary for tumorigenesis. CONCLUSIONS HSP90 inhibition showing significant antitumor activity against NF2-related tumor cells in vitro and in vivo represents a promising option for novel NF2 therapies.
Collapse
Affiliation(s)
- Karo Tanaka
- Center for Neural Tumor Research and Section on Genetics of Hereditary Ear Disorders, House Research Institute, University of California, Los Angeles, CA 90057, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Curic S, Wu Y, Shan B, Schaaf C, Utpadel D, Lange M, Kuhlen D, Perone MJ, Arzt E, Stalla GK, Renner U. Curcumin acts anti-proliferative and pro-apoptotic in human meningiomas. J Neurooncol 2013; 113:385-96. [DOI: 10.1007/s11060-013-1148-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 05/06/2013] [Indexed: 12/25/2022]
|
27
|
Angelo LS, Maxwell DS, Wu JY, Sun D, Hawke DH, McCutcheon IE, Slopis JM, Peng Z, Bornmann WG, Kurzrock R. Binding partners for curcumin in human schwannoma cells: biologic implications. Bioorg Med Chem 2013; 21:932-9. [PMID: 23294827 DOI: 10.1016/j.bmc.2012.12.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 11/29/2012] [Accepted: 12/06/2012] [Indexed: 02/03/2023]
Abstract
Curcumin (diferuloylmethane) is a potent anti-inflammatory and anti-tumorigenic agent that has shown preclinical activity in diverse cancers. Curcumin up-regulates heat shock protein 70 (hsp70) mRNA in several different cancer cell lines. Hsp70 contributes to an escape from the apoptotic effects of curcumin by several different mechanisms including prevention of the release of apoptosis inducing factor from the mitochondria and inhibition of caspases 3 and 9. Previously we showed that the combination of curcumin plus a heat shock protein inhibitor was synergistic in its down-regulation of the proliferation of a human schwannoma cell line (HEI-193) harboring an NF2 mutation, possibly because curcumin up-regulated hsp70, which also binds merlin, the NF2 gene product. In order to determine if curcumin also interacts directly with hsp70 and to discover other binding partners of curcumin, we synthesized biotinylated curcumin (bio-curcumin) and treated HEI-193 schwannoma cells. Cell lysates were prepared and incubated with avidin-coated beads. Peptides pulled down from this reaction were sequenced and it was determined that biotinylated curcumin bound hsp70, hsp90, 3-phosphoglycerate dehydrogenase, and a β-actin variant. These binding partners may serve to further elucidate the underlying mechanisms of curcumin's actions.
Collapse
Affiliation(s)
- Laura S Angelo
- Department of Investigational Cancer Therapeutics, (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Development of drug treatments for neurofibromatosis type 2-associated vestibular schwannoma. Curr Opin Otolaryngol Head Neck Surg 2013; 20:372-9. [PMID: 22931905 DOI: 10.1097/moo.0b013e328357d2ee] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To review the discoveries in molecular pathophysiology contributing to the development of neurofibromatosis type 2 (NF2)-associated vestibular schwannomas and the recent experiences with drug therapies for these tumors. The review includes discussion of diagnostic criteria for NF2, populations to clinically consider for drug therapy and drug targets currently under consideration for NF2. RECENT FINDINGS Increased insight into the complex pathways that underlie both the genetic syndrome of NF2 and the specific pathogenesis of vestibular schwannomas has highlighted multiple potential therapeutic targets. These discoveries have been translated into clinical trials with some early promising results. Inhibition of angiogenesis as well as regulation of mammalian target of rapamycin and the epidermal growth factor receptor family of receptors are the focus of current clinical investigations. SUMMARY Although a great deal of work is ongoing to understand the multiple effects of the lack of the regulating protein Merlin on tumorgenesis in patients with NF2, advances are ongoing with clinical therapeutics. There is cause for enthusiasm based on recent results with antiangiogenesis therapy in select patients with NF2 and progressive vestibular schwannomas; however, awareness of the notable risks and limitations of therapies currently in development is required.
Collapse
|
29
|
D'Aguanno S, D'Agnano I, De Canio M, Rossi C, Bernardini S, Federici G, Urbani A. Shotgun proteomics and network analysis of neuroblastoma cell lines treated with curcumin. MOLECULAR BIOSYSTEMS 2012; 8:1068-77. [DOI: 10.1039/c2mb05498a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|