1
|
Hu Z, Qin Z, Xie J, Qu Y, Yin L. Cannabidiol and its application in the treatment of oral diseases: therapeutic potentials, routes of administration and prospects. Biomed Pharmacother 2024; 176:116271. [PMID: 38788594 DOI: 10.1016/j.biopha.2024.116271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/27/2024] [Accepted: 02/07/2024] [Indexed: 05/26/2024] Open
Abstract
Cannabidiol (CBD), one of the most important active ingredients in cannabis, has been reported to have some pharmacological effects such as antibacterial and analgesic effects, and to have therapeutic potential in the treatment of oral diseases such as oral cancer, gingivitis and periodontal diseases. However, there is a lack of relevant systematic research and reviews. Therefore, based on the etiology and clinical symptoms of several common oral diseases, this paper focuses on the therapeutic potential of CBD in periodontal diseases, pulp diseases, oral mucosal diseases, oral cancer and temporomandibular joint diseases. The pharmacological effects of CBD and the distribution and function of its receptors in the oral cavity are also summarized. In order to provide reference for future research and further clinical application of CBD, we also summarize several possible routes of administration and corresponding characteristics. Finally, the challenges faced while applying CBD clinically and possible solutions are discussed, and we also look to the future.
Collapse
Affiliation(s)
- Zonghao Hu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Zishun Qin
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Jinhong Xie
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Yue Qu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Lihua Yin
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Seegobin M, Logan SR, Emery RJN, Brunetti CR. Cytokinins Reduce Viral Replication and Alter Plaque Morphology of Frog Virus 3 In Vitro. Viruses 2024; 16:826. [PMID: 38932119 PMCID: PMC11209418 DOI: 10.3390/v16060826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Cytokinins (CKs) are a group of N6-substituted signaling molecules whose biosynthesis and metabolism have been documented in all kingdoms of life, including vertebrates. While their biological relevance in vertebrate systems continues to be elucidated, they have broadly been documented with therapeutic effects in exogenous applications. In this study, we evaluated the virostatic potential of four types of CKs including, N6-isopentenyladenine (iP), N6-isopentenyladenosine (iPR), N6-isopentenyladenosine-5'monophosphate (iPMP), and 2-methylthiol-N6-isopentenyladenosine (2MeSiPR) against the ranavirus type species, frog virus 3 (FV3). Following concurrent treatment and infection, iP and iPR reduced viral replication by 33.8% and 59.6%, respectively, in plaque formation assays. A decrease in viral replication was also observed when CK exposure was limited to 12 h prior to infection, where iP and iPR reduced viral replication by 31% and 23.75%, respectively. Treatment with iP and iPR was also marked by 48% and 60% decreases in viral load over 72 h, respectively, as measured in single step growth curves. Plaque morphology was altered in vitro, as iP and iPR treatment increased plaque area by 83% and 112% with lytic zone formation also becoming more prevalent in corresponding treatments. Treatment with iPMP and 2MeSiPR resulted in no effect on viral kinetics in vitro. The results of this study are the first to provide evidence of CK antiviral activity against a DNA virus and highlight the importance of their structure for therapeutic investigations.
Collapse
Affiliation(s)
| | | | | | - Craig R. Brunetti
- Department of Biology, Trent University, 1600 West Bank Drive, Peterborough, ON K9J 0G2, Canada; (M.S.); (S.R.L.); (R.J.N.E.)
| |
Collapse
|
3
|
Pagano C, Coppola L, Navarra G, Avilia G, Savarese B, Torelli G, Bruzzaniti S, Piemonte E, Galgani M, Laezza C, Bifulco M. N6-isopentenyladenosine inhibits aerobic glycolysis in glioblastoma cells by targeting PKM2 expression and activity. FEBS Open Bio 2024; 14:843-854. [PMID: 38514913 PMCID: PMC11073503 DOI: 10.1002/2211-5463.13766] [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: 09/18/2023] [Revised: 12/20/2023] [Accepted: 01/08/2024] [Indexed: 03/23/2024] Open
Abstract
Glioblastoma (GBM) is a primary tumor in the central nervous system with poor prognosis. It exhibits elevated glucose uptake and lactate production. This metabolic state of aerobic glycolysis is known as the Warburg effect. N6-isopentenyladenosine (iPA), a natural cytokine modified with an isopentenyl moiety derived from the mevalonate pathway, has well-established anti-tumor activity. It inhibits cell proliferation in glioma cells, inducing cell death by apoptosis and/or necroptosis. In the present study, we found that iPA inhibits aerobic glycolysis in unmodified U87MG cells and in the same cell line engineered to over-express wild-type epidermal growth factor receptor (EGFR) or EGFR variant III (vIII), as well as in a primary GBM4 patient-derived cell line. The detection of glycolysis showed that iPA treatment suppressed ATP and lactate production. We also evaluated the response of iPA treatment in normal human astrocyte primary cells, healthy counterpart cells of the brain. Aerobic glycolysis in treated normal human astrocyte cells did not show significant changes compared to GBM cells. To determine the mechanism of iPA action on aerobic glycolysis, we investigated the expression of certain enzymes involved in this metabolic pathway. We observed that iPA reduced the expression of pyruvate kinase M2 (PKM2), which plays a key role in the regulation of aerobic glycolysis, promoting tumor cell proliferation. The reduction of PKM2 expression is a result of the inhibition of the inhibitor of nuclear factor kappa-B kinase subunit, beta/nuclear factor-kappa B pathway upon iPA treatment. In conclusion, these experimental results show that iPA may inhibit aerobic glycolysis of GBM in stabilized cell lines and primary GBM cells by targeting the expression and activity of PKM2.
Collapse
Affiliation(s)
- Cristina Pagano
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples ‘Federico II’NaplesItaly
| | - Laura Coppola
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples ‘Federico II’NaplesItaly
| | - Giovanna Navarra
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples ‘Federico II’NaplesItaly
| | - Giorgio Avilia
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples ‘Federico II’NaplesItaly
| | - Beatrice Savarese
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples ‘Federico II’NaplesItaly
| | - Giovanni Torelli
- Neurosurgery Unit A.O. San Giovanni di Dio e Ruggi d' Aragona – Salerno's School of Medicine Largo Città di IppocrateSalernoItaly
| | - Sara Bruzzaniti
- Institute of Endocrinology and Experimental Oncology (IEOS)National Research Council (CNR)NaplesItaly
| | - Erica Piemonte
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples ‘Federico II’NaplesItaly
| | - Mario Galgani
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples ‘Federico II’NaplesItaly
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology (IEOS)National Research Council (CNR)NaplesItaly
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples ‘Federico II’NaplesItaly
| |
Collapse
|
4
|
Lopardo V, Montella F, Esposito RM, Zannella C, Aliberti SM, Capunzo M, Franci G, Puca AA, Ciaglia E. SARS-CoV-2 Lysate Stimulation Impairs the Release of Platelet-like Particles and Megakaryopoiesis in the MEG-01 Cell Line. Int J Mol Sci 2023; 24:4723. [PMID: 36902151 PMCID: PMC10003077 DOI: 10.3390/ijms24054723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023] Open
Abstract
SARS-CoV-2 infection causes a considerable inflammatory response coupled with impaired platelet reactivity, which can lead to platelet disorders recognized as negative prognostic factors in COVID-19 patients. The virus may cause thrombocytopenia or thrombocytosis during the different disease stages by destroying or activating platelets and influencing platelet production. While it is known that several viruses can impair megakaryopoiesis by generating an improper production and activation of platelets, the potential involvement of SARS-CoV-2 in affecting megakaryopoiesis is poorly understood. To this purpose, we explored, in vitro, the impact of SARS-CoV-2 stimulation in the MEG-01 cell line, a human megakaryoblastic leukemia cell line, considering its spontaneous capacity of releasing platelet-like particles (PLPs). We interrogated the effect of heat-inactivated SARS-CoV-2 lysate in the release of PLPs and activation from MEG-01, the signaling pathway influenced by SARS-CoV-2, and the functional effect on macrophagic skewing. The results highlight the potential influence of SARS-CoV-2 in the early stages of megakaryopoiesis by enhancing the production and activation of platelets, very likely due to the impairment of STATs signaling and AMPK activity. Overall, these findings provide new insight into the role of SARS-CoV-2 in affecting megakaryocyte-platelet compartment, possibly unlocking another avenue by which SARS-CoV-2 moves.
Collapse
Affiliation(s)
- Valentina Lopardo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvatore Allende, 84081 Baronissi, Italy
| | - Francesco Montella
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvatore Allende, 84081 Baronissi, Italy
| | - Roberta Maria Esposito
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvatore Allende, 84081 Baronissi, Italy
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Silvana Mirella Aliberti
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvatore Allende, 84081 Baronissi, Italy
| | - Mario Capunzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvatore Allende, 84081 Baronissi, Italy
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvatore Allende, 84081 Baronissi, Italy
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Annibale Alessandro Puca
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvatore Allende, 84081 Baronissi, Italy
- Cardiovascular Research Unit, IRCCS MultiMedica, 20138 Milan, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvatore Allende, 84081 Baronissi, Italy
| |
Collapse
|
5
|
Pagano C, Coppola L, Navarra G, Avilia G, Bruzzaniti S, Piemonte E, Galgani M, Della Monica R, Chiariotti L, Cuomo M, Buonaiuto M, Torelli G, Caiazzo P, Laezza C, Bifulco M. N6-Isopentenyladenosine Impairs Mitochondrial Metabolism through Inhibition of EGFR Translocation on Mitochondria in Glioblastoma Cells. Cancers (Basel) 2022; 14:cancers14246044. [PMID: 36551529 PMCID: PMC9776489 DOI: 10.3390/cancers14246044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive malignant brain tumor and is poorly susceptible to cytotoxic therapies. Amplification of the epidermal growth factor receptor (EGFR) and deletion of exons 2 to 7, which generates EGFR variant III (vIII), are the most common molecular alterations of GBMs that contribute to the aggressiveness of the disease. Recently, it has been shown that EGFR/EGFRvIII-targeted inhibitors enhance mitochondrial translocation by causing mitochondrial accumulation of these receptors, promoting the tumor drug resistance; moreover, they negatively modulate intrinsic mitochondria-mediated apoptosis by sequestering PUMA, leading to impaired apoptotic response in GBM cells. N6-isopentenyladenosine (i6A or iPA), a cytokinin consisting of an adenosine linked to an isopentenyl group deriving from the mevalonate pathway, has antiproliferative effects on numerous tumor cells, including GBM cells, by inducing cell death in vitro and in vivo. Here, we observed that iPA inhibits the mitochondrial respiration in GBM cells by preventing the translocation of EGFR/EGFRvIII to the mitochondria and allowing PUMA to interact with them by promoting changes in mitochondrial activity, thus playing a critical role in cell death. Our findings clearly demonstrate that iPA interferes with mitochondrial bioenergetic capacity, providing a rationale for an effective strategy for treating GBM.
Collapse
Affiliation(s)
- Cristina Pagano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
| | - Laura Coppola
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
| | - Giovanna Navarra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
| | - Giorgio Avilia
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
| | - Sara Bruzzaniti
- Department of Biology, University of Naples “Federico II”, 80126 Naples, Italy
- Institute of Endocrinology and Experimental Oncology (IEOS), National Research Council (CNR), 80125 Naples, Italy
| | - Erica Piemonte
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
| | - Mario Galgani
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
- Institute of Endocrinology and Experimental Oncology (IEOS), National Research Council (CNR), 80125 Naples, Italy
| | - Rosa Della Monica
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
- CEINGE—Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80131 Naples, Italy
| | - Lorenzo Chiariotti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
- CEINGE—Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80131 Naples, Italy
| | - Mariella Cuomo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
- CEINGE—Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80131 Naples, Italy
| | - Michela Buonaiuto
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
- CEINGE—Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80131 Naples, Italy
| | - Giovanni Torelli
- Neurosurgery Unit A.O. San Giovanni di Dio e Ruggi d’ Aragona, Salerno’s School of Medicine Largo Città di Ippocrate, 84131 Salerno, Italy
- Osservatorio Oncologico, 84091 Battipaglia, Italy
| | | | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology (IEOS), National Research Council (CNR), 80125 Naples, Italy
- Correspondence: or (C.L.); (M.B.)
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy
- Correspondence: or (C.L.); (M.B.)
| |
Collapse
|
6
|
Proto MC, Fiore D, Piscopo C, Laezza C, Bifulco M, Gazzerro P. Modified Adenosines Sensitize Glioblastoma Cells to Temozolomide by Affecting DNA Methyltransferases. Front Pharmacol 2022; 13:815646. [PMID: 35559231 PMCID: PMC9086827 DOI: 10.3389/fphar.2022.815646] [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: 11/15/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal primary malignant brain tumor, and due to its unique features, its management is certainly one of the most challenging ones among all cancers. N6-isopentenyladenosine (IPA) and its analog N6-benzyladenosine (N6-BA) are modified nucleosides endowed with potent antitumor activity on different types of human cancers, including GBM. Corroborating our previous finding, we demonstrated that IPA and N6-BA affect GBM cell line proliferation by modulating the expression of the F-box WD repeat domain-containing-7 (FBXW7), a tumor suppressor with a crucial role in the turnover of many proteins, such as SREBPs and Mcl1, involved in malignant progression and chemoresistance. Luciferase assay revealed that IPA-mediated upregulation of FBXW7 translates in transcriptional inactivation of its oncogenic substrates (Myc, NFkB, or HIF-1α). Moreover, downregulating MGMT expression, IPA strongly enhances the killing effect of temozolomide (TMZ), producing a favorable sensitizing effect starting from a concentration range much lower than TMZ EC50. Through DNA methyltransferase (DNMT) activity assay, analysis of the global DNA methylation, and the histone modification profiles, we demonstrated that the modified adenosines behave similar to 5-AZA-dC, known DNMT inhibitor. Overall, our results provide new perspectives for the first time, suggesting the modified adenosines as epigenetic tools able to improve chemo- and radiotherapy efficacy in glioblastoma and potentially other cancers.
Collapse
Affiliation(s)
| | - Donatella Fiore
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Chiara Piscopo
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, Naples, Italy
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | | |
Collapse
|
7
|
Transfer of the longevity-associated variant of BPIFB4 gene rejuvenates immune system and vasculature by a reduction of CD38 + macrophages and NAD + decline. Cell Death Dis 2022; 13:86. [PMID: 35087020 PMCID: PMC8792139 DOI: 10.1038/s41419-022-04535-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/20/2021] [Accepted: 01/13/2022] [Indexed: 01/10/2023]
Abstract
As we age, our body experiences chronic, systemic inflammation contributing to the morbidity and mortality of the elderly. The senescent immune system has been described to have a causal role in driving systemic aging and therefore may represent a key therapeutic target to prevent pathological consequences associated with aging and extend a healthy lifespan. Previous studies from our group associated a polymorphic haplotype variant in the BPIFB4 gene (LAV-BPIFB4) with exceptional longevity. Transfer of the LAV-BPIFB4 in preclinical models halted the progression of cardiovascular diseases (CVDs) and frailty by counterbalancing chronic inflammation. In the present study, we aimed to delineate the action of systemic adeno-associated viral vector-mediated LAV-BPIFB4 gene transfer (AAV-LAV-BPIFB4) on the deleterious age-related changes of the immune system and thereby the senescence-associated events occurring in C57BL/6J mice aged 26 months. Our in vivo data showed that 26-months-old mice had a higher frequency of CD45+SA-beta Gal+ immune cells in peripheral blood than young (4-months-old) C57BL/6J mice. Notably, AAV-LAV-BPIFB4 gene transfer in aged mice reduced the pool of peripheral immunosenescent cells that were shown to be enriched in the spleen. In addition, the proper tuning of the immune secretory phenotype (IL1βlow, IL6low, IL10high) associated with a significant reduction in SA-beta Gal-positive area of aorta from AAV-LAV treated mice. At the functional level, the reduction of senescence-associated inflammation ensured sustained NAD+ levels in the plasma of AAV-LAV-BPIFB4 old mice by preventing the NADase CD38 increase in F4/80+ tissue-resident macrophages and Ly6Chigh pro-inflammatory monocytes of the spleen and bone marrow. Finally, to validate the clinical implication of our findings, we showed that Long-living-individuals (LLIs, >95 years), which delay CVDs onset, especially if LAV-carriers, were characterized by high NAD+ levels. In conclusion, the new senotherapeutic action of LAV-BPIFB4 may offer a valuable therapeutic tool to control aging and reduce the burden of its pathophysiological disorders, such as CVDs.
Collapse
|
8
|
Adami R, Bottai D. S-adenosylmethionine tRNA modification: unexpected/unsuspected implications of former/new players. Int J Biol Sci 2020; 16:3018-3027. [PMID: 33061813 PMCID: PMC7545696 DOI: 10.7150/ijbs.49302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
S-adenosylmethionine supplies methyl groups to many acceptors, including lipids, proteins, RNA, DNA, and a wide range of small molecules. It acts as the precursor in the biosynthesis of metal ion chelating compounds, such as nicotianamine and phytosiderophores, of the polyamines spermidine and spermine and of some plant hormones. Finally, it is the source of catalytic 5′-deoxyadenosyl radicals. Radical S-adenosylmethionine (SAM) enzymes (RS) represent one of the most abundant groups (more than 100,000) of enzymes, exerting a plethora of biological functions, some of which are still unknown. In this work, we will focus on two RS: CDK5RAP1 and CDKAL1, both of which are involved in tRNA modifications that result in important tRNA folding and stability and in maintaining high translational fidelity. Based on this crucial role, their impairment can be important in the development of different human diseases.
Collapse
Affiliation(s)
- Raffaella Adami
- Department of Health Science University of Milan via A. di Rudinì 8 20142 Milan
| | - Daniele Bottai
- Department of Health Science University of Milan via A. di Rudinì 8 20142 Milan
| |
Collapse
|
9
|
Oshchepkov MS, Kalistratova AV, Savelieva EM, Romanov GA, Bystrova NA, Kochetkov KA. Natural and synthetic cytokinins and their applications in biotechnology, agrochemistry and medicine. RUSSIAN CHEMICAL REVIEWS 2020. [DOI: 10.1070/rcr4921] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The review is devoted to cytokinins — classical plant hormones known for more than six decades. Nevertheless, different aspects of the action of cytokinins are still being investigated. Relevant studies produced interesting, often unexpected, results, which cast doubt on the old paradigms and open new prospects for the use of these phytohormones. Particular attention is given to recent advances in the applications of natural cytokinins and their synthetic analogues in biotechnology, agriculture, medicine and cosmetics. The chemical synthesis, properties and the possible use of artificial cytokinins are considered in detail. The review is aimed at researchers interested in the development and applications of new biologically active compounds with a wide spectrum of action on diverse biological objects, from plants to humans.
The bibliography includes 233 references.
Collapse
|
10
|
The longevity-associated variant of BPIFB4 improves a CXCR4-mediated striatum-microglia crosstalk preventing disease progression in a mouse model of Huntington's disease. Cell Death Dis 2020; 11:546. [PMID: 32683420 PMCID: PMC7368858 DOI: 10.1038/s41419-020-02754-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 01/10/2023]
Abstract
The longevity-associated variant (LAV) of the bactericidal/permeability-increasing fold-containing family B member 4 (BPIFB4) has been found significantly enriched in long-living individuals. Neuroinflammation is a key player in Huntington's disease (HD), a neurodegenerative disorder caused by neural death due to expanded CAG repeats encoding a long polyglutamine tract in the huntingtin protein (Htt). Herein, we showed that striatal-derived cell lines with expanded Htt (STHdh Q111/111) expressed and secreted lower levels of BPIFB4, when compared with Htt expressing cells (STHdh Q7/7), which correlated with a defective stress response to proteasome inhibition. Overexpression of LAV-BPIFB4 in STHdh Q111/111 cells was able to rescue both the BPIFB4 secretory profile and the proliferative/survival response. According to a well-established immunomodulatory role of LAV-BPIFB4, conditioned media from LAV-BPIFB4-overexpressing STHdh Q111/111 cells were able to educate Immortalized Human Microglia-SV40 microglial cells. While STHdh Q111/111 dying cells were ineffective to induce a CD163 + IL-10high pro-resolving microglia compared to normal STHdh Q7/7, LAV-BPIFB4 transduction promptly restored the central immune control through a mechanism involving the stromal cell-derived factor-1. In line with the in vitro results, adeno-associated viral-mediated administration of LAV-BPIFB4 exerted a CXCR4-dependent neuroprotective action in vivo in the R6/2 HD mouse model by preventing important hallmarks of the disease including motor dysfunction, body weight loss, and mutant huntingtin protein aggregation. In this view, LAV-BPIFB4, due to its pleiotropic ability in both immune compartment and cellular homeostasis, may represent a candidate for developing new treatment for HD.
Collapse
|
11
|
Grimaldi M, Randino R, Ciaglia E, Scrima M, Buonocore M, Stillitano I, Abate M, Covelli V, Tosco A, Gazzerro P, Bifulco M, Rodriquez M, D'Ursi AM. NMR for screening and a biochemical assay: Identification of new FPPS inhibitors exerting anticancer activity. Bioorg Chem 2019; 98:103449. [PMID: 32057422 DOI: 10.1016/j.bioorg.2019.103449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/01/2019] [Accepted: 11/14/2019] [Indexed: 01/14/2023]
Abstract
Farnesyl pyrophosphate synthase (FPPS) is a crucial enzyme for the synthesis of isoprenoids and the key target of nitrogen-containing bisphosphonates (N-BPs). N-BPs are potent and selective FPPS inhibitors that are used in the treatment of bone-related diseases, but have poor pharmacokinetic properties. Given the key role played by FPPS in many cancer-related pathways and the pharmacokinetic limits of N-BPs, hundreds of molecules have been screened to identify new FPPS inhibitors characterized by improved drug-like properties that are useful for broader therapeutic applications in solid, non-skeletal tumours. We have previously shown that N6-isopentenyladenosine (i6A) and its related compound N6-benzyladenosine (2) exert anti-glioma activity by interfering with the mevalonate pathway and inhibiting FPPS. Here, we report the design and synthesis of a panel of N6-benzyladenosine derivatives (compounds 2a-m) incorporating different chemical moieties on the benzyl ring. Compounds 2a-m show in vitro antiproliferative activity in U87MG glioma cells and, analogous to the bisphosphonate FPPS inhibitors, exhibit immunogenic properties in ex vivo γδ T cells from stimulated peripheral blood mononuclear cells (PBMCs). Using saturation transfer difference (STD) and quantitative 1H nuclear magnetic resonance (NMR) experiments, we found that 2f, the N6-benzyladenosine analogue that includes a tertbutyl moiety in the para position of the benzyl ring, is endowed with increased FPPS binding and inhibition compared to the parent compounds i6A and 2. N6-benzyladenosine derivatives, characterized by structural features that are significantly different from those of N-BPs, have been confirmed to be promising chemical scaffolds for the development of non N-BP FPPS inhibitors, exerting combined cytotoxic and immunostimulatory activities.
Collapse
Affiliation(s)
- Manuela Grimaldi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, V.le J.F. Kennedy 54 - Pad. 20 Mostra d'Oltremare, 80125 Naples, Italy
| | - Rosario Randino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081 Baronissi, Salerno, Italy
| | - Mario Scrima
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Michela Buonocore
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Ilaria Stillitano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Mario Abate
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081 Baronissi, Salerno, Italy
| | - Verdiana Covelli
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Alessandra Tosco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini, 80131 Naples, Italy; Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081 Baronissi, Salerno, Italy
| | - Manuela Rodriquez
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Anna Maria D'Ursi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy.
| |
Collapse
|
12
|
Salviati E, Ciaglia E, Sommella E, Montella F, Bertamino A, Ostacolo C, Parrino B, Rubino R, Vecchione C, Puca A, Novellino E, Campiglia P. Immunomodulatory activity of Humulus lupulus bitter acids fraction: Enhancement of natural killer cells function by NKp44 activating receptor stimulation. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
13
|
Ciaglia E, Montella F, Trucillo P, Ciardulli M, Di Pietro P, Amodio G, Remondelli P, Vecchione C, Reverchon E, Maffulli N, Puca A, Della Porta G. A bioavailability study on microbeads and nanoliposomes fabricated by dense carbon dioxide technologies using human-primary monocytes and flow cytometry assay. Int J Pharm 2019; 570:118686. [DOI: 10.1016/j.ijpharm.2019.118686] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/06/2019] [Accepted: 09/07/2019] [Indexed: 12/19/2022]
|
14
|
Seegobin M, Kisiala A, Noble A, Kaplan D, Brunetti C, Emery RJN. Canis familiaris tissues are characterized by different profiles of cytokinins typical of the tRNA degradation pathway. FASEB J 2018; 32:fj201800347. [PMID: 29894666 DOI: 10.1096/fj.201800347] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Cytokinins (CKs) are a group of phytohormones essential to plant growth and development. The presence of these N6-modified adenine derivatives has also been documented in other groups of organisms, including bacteria, fungi, and insects. Thus far, however, only a single CK, N6-(Δ2-isopentenyl) adenine-9-riboside (iPR), has been identified in mammals. In plants, the nucleotide form of isopentenyladenine [iPR (either mono-, di-, or tri-) phosphate (iPRP)] is the first form of CK synthesized, and it is further modified to produce other CK types. To determine if a similar biosynthesis pathway exists in mammals, we tested for the presence of 27 CKs in a wide selection of canine organs using HPLC electrospray ionization-tandem mass spectrometry. Seven forms of CK were detected in the majority of the analyzed samples, including iPR, iPRP, cis-zeatin-9-riboside, cis-zeatin-9-riboside-5' (either mono-, di-, or triphosphate), 2-methylthio-N6-isopentenyladenine, 2-methylthio-N6-isopentenyladenosine, and 2-methylthio-zeatin. Total CK concentrations ranged from 1.96 pmol/g fresh weight (adrenal glands) to 1.40 × 103 pmol/g fresh weight (thyroid). The results of this study provide evidence that mammalian cells, like plant cells, can synthesize and process a diverse set of CKs including cis- and methylthiol-type CKs.-Seegobin, M., Kisiala, A., Noble, A., Kaplan, D., Brunetti, C., Emery, R. J. N. Canis familiaris tissues are characterized by different profiles of cytokinins typical of tRNA degradation pathway.
Collapse
Affiliation(s)
- Mark Seegobin
- Department of Biology, Trent University, Peterborough, Ontario, Canada
| | - Anna Kisiala
- Department of Biology, Trent University, Peterborough, Ontario, Canada
| | | | - David Kaplan
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Craig Brunetti
- Department of Biology, Trent University, Peterborough, Ontario, Canada
| | - R J Neil Emery
- Department of Biology, Trent University, Peterborough, Ontario, Canada
| |
Collapse
|
15
|
Santoro A, Ciaglia E, Nicolin V, Pescatore A, Prota L, Capunzo M, Ursini MV, Nori SL, Bifulco M. The isoprenoid end product N6-isopentenyladenosine reduces inflammatory response through the inhibition of the NFκB and STAT3 pathways in cystic fibrosis cells. Inflamm Res 2017; 67:315-326. [PMID: 29230506 PMCID: PMC5843674 DOI: 10.1007/s00011-017-1123-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 12/03/2017] [Accepted: 12/06/2017] [Indexed: 12/26/2022] Open
Abstract
Objective N6-isopentenyladenosine (iPA) is an intermediate of the mevalonate pathway that exhibits various anti-cancer effects. However, studies on its anti-inflammatory activity are scarce and underlying molecular mechanisms are unknown. Therefore, we aimed to investigate the ability of iPA to exert anti-inflammatory effects in the human cystic fibrosis (CF) cell model of exacerbated inflammation. Materials and methods TNFα-stimulated CF cells CuFi-1 and its normal counterpart NuLi-1 were pre-treated with increasing concentrations of iPA and cell viability and proliferation were assessed by MTT and BrdU assays. The effect of iPA on IL-8 and RANTES secretion was determined by ELISA, and the activation and expression of signaling molecules and selenoproteins were studied by Western blot. To assess the direct effect of iPA on NFκB activity, luciferase assay was performed on TNFα-stimulated HEK293/T cells transfected with a NFκB reporter plasmid. Results We demonstrated for the first time that iPA prevents IL-8 and RANTES release in TNFα-stimulated CF cells and this effect is mediated by increasing the expression of the direct NFκB inhibitor IκBα and decreasing the levels of STAT3. Consistent with this, we showed that iPA inhibited TNFα-mediated NFκB activation in HEK/293T cells. Finally, we also found that iPA improved the levels of glutathione peroxidase 1 and thioredoxin reductase 1 only in CF cells suggesting its ability to maintain sufficient expression of these anti-oxidant selenoproteins. Conclusions Our findings indicate that iPA can exert anti-inflammatory activity especially in the cases of excessive inflammatory response as in CF.
Collapse
Affiliation(s)
- Antonietta Santoro
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Via S. Allende, 84081, Baronissi, Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Via S. Allende, 84081, Baronissi, Salerno, Italy
| | - Vanessa Nicolin
- Clinical Department of Medical, Surgical and Health Science, University of Trieste, Strada di Fiume 447, 34149, Trieste, Italy
| | - Alessandra Pescatore
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso" CNR, Via P. Castellino, 80131, Naples, Italy
| | - Lucia Prota
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Via S. Allende, 84081, Baronissi, Salerno, Italy
| | - Mario Capunzo
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Via S. Allende, 84081, Baronissi, Salerno, Italy
| | - Matilde V Ursini
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso" CNR, Via P. Castellino, 80131, Naples, Italy
| | - Stefania L Nori
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Via S. Allende, 84081, Baronissi, Salerno, Italy.
| | - Maurizio Bifulco
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Via S. Allende, 84081, Baronissi, Salerno, Italy. .,Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini, 80131, Naples, Italy.
| |
Collapse
|
16
|
Ranieri R, Ciaglia E, Amodio G, Picardi P, Proto MC, Gazzerro P, Laezza C, Remondelli P, Bifulco M, Pisanti S. N6-isopentenyladenosine dual targeting of AMPK and Rab7 prenylation inhibits melanoma growth through the impairment of autophagic flux. Cell Death Differ 2017; 25:353-367. [PMID: 29027991 DOI: 10.1038/cdd.2017.165] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/03/2017] [Accepted: 08/06/2017] [Indexed: 12/12/2022] Open
Abstract
Targeting the autophagic process is considered a promising therapeutic strategy in cancer since a great number of tumors, including melanoma, show high basal levels of protective autophagy that contributes to tumor progression and chemoresistance. Here, exploiting both in vitro and in vivo approaches, we identified N6-isopentenyladenosine (iPA), an end product of the mevalonate pathway, as a novel autophagy inhibitor with an interesting anti-melanoma activity. iPA, after being phosphorylated by adenosine kinase into 5'-iPA-monophosphate, induces autophagosome accumulation through AMPK activation, measured by increased fluorescent GFP-LC3 puncta and enhanced conversion into the lipidated autophagosome-associated LC3-II. However, at a later stage iPA blocks the autophagic flux monitored by p62 accumulation, Luciferase reporter-based assay for LC3 turnover in living cells and fluorescence of a tandem RFP-GFP-LC3 construct. Impaired autophagic flux is due to the block of autophagosome-lysosome fusion through the defective localization and function of Rab7, whose prenylation is inhibited by iPA, resulting in a net inhibition of autophagy completion that finally leads to melanoma apoptotic cell death. AMPK silencing prevents apoptosis upon iPA treatment, whereas basal autophagosome turnover is still inhibited due to unprenylated Rab7. These results strongly support the advantage of targeting autophagy for therapeutic gain in melanoma and provide the preclinical rational to further investigate the antitumor action of iPA, able to coordinately induce autophagosome accumulation and inhibit the autophagic flux, independently targeting AMPK and Rab7 prenylation. This property may be particularly useful for the selective killing of tumors, like melanoma, that frequently develop chemotherapy resistance due to protective autophagy activation.
Collapse
Affiliation(s)
- Roberta Ranieri
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Giuseppina Amodio
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Paola Picardi
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | | | | | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology (IEOS), Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Paolo Remondelli
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Maurizio Bifulco
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Simona Pisanti
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| |
Collapse
|
17
|
Ciaglia E, Laezza C, Abate M, Pisanti S, Ranieri R, D'alessandro A, Picardi P, Gazzerro P, Bifulco M. Recognition by natural killer cells of N6-isopentenyladenosine-treated human glioma cell lines. Int J Cancer 2017; 142:176-190. [PMID: 28884474 DOI: 10.1002/ijc.31036] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 08/09/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022]
Abstract
Cancer cell stress induced by cytotoxic agents promotes antitumor immune response. Here, we observed that N6-isopentenyladenosine (iPA), an isoprenoid modified adenosine with a well established anticancer activity, was able to induce a significant upregulation of cell surface expression of natural killer (NK) cell activating receptor NK Group 2 member D (NKG2D) ligands on glioma cells in vitro and xenografted in vivo. Specifically suboptimal doses of iPA (0.1 and 1 µM) control the selective upregulation of UL16-binding protein 2 on p53wt-expressing U343MG and that of MICA/B on p53mut-expressing U251MG cells. This event made the glioblastoma cells a potent target for NK cell-mediated recognition through a NKG2D restricted mechanism. p53 siRNA-mediated knock-down and pharmacological inhibition (pifithrin-α), profoundly prevented the iPA action in restoring the immunogenicity of U343MG cells through a mechanism that is dependent upon p53 status of malignancy. Furthermore, accordingly to the preferential recognition of senescent cells by NK cells, we found that iPA treatment was critical for glioma cells entry in premature senescence through the induction of S and G2/M phase arrest. Collectively, our results indicate that behind the well established cytotoxic and antiangiogenic effects, iPA can also display an immune-mediated antitumor activity. The indirect engagement of the innate immune system and its additional activity in primary derived patient's glioma cell model (GBM17 and GBM37), fully increase its translational relevance and led to the exploitation of the isoprenoid pathway for a valid therapeutic intervention in antiglioma research.
Collapse
Affiliation(s)
- Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, Baronissi Salerno, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, Via Pansini 5, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Via Pansini, Naples, Italy
| | - Mario Abate
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, Baronissi Salerno, Italy
| | - Simona Pisanti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, Baronissi Salerno, Italy
| | - Roberta Ranieri
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, Baronissi Salerno, Italy
| | - Alba D'alessandro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano Salerno, Italy
| | - Paola Picardi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano Salerno, Italy.,Axxam Spa OpenZone - via A. Meucci, Bresso, Milano, Italy
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano Salerno, Italy
| | - Maurizio Bifulco
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, Baronissi Salerno, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Via Pansini, Naples, Italy
| |
Collapse
|
18
|
申 九, 熊 共, 郑 启, 张 宏, 洪 再. 自然杀伤细胞抑制肝癌肺转移. Shijie Huaren Xiaohua Zazhi 2017; 25:2028-2038. [DOI: 10.11569/wcjd.v25.i22.2028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
目的 研究自然杀伤(natural killer, NK)细胞对肝癌的抑制作用, 为临床应用提供实验依据.
方法 从人外周血分离培养及鉴定NK细胞. 在体外, 研究NK细胞抑制肝癌细胞的增殖、迁徙、转移. 在体内, 检测NK细胞在裸鼠肝脏存活情况. 利用人肝癌组织裸鼠肝脏原位移植模型来评估NK细胞在体内对肝癌生长、转移的抑制功能. 通过检测NK细胞活化受体、NKB1、穿孔素和颗粒酶的表达情况来评估白介素(interleukin, IL)-2对NK细胞免疫功能的刺激作用.
结果 采用密度梯度法可以获取较大量的外周血单个核细胞, 且能够从中分离到高活力的NK细胞. NK细胞经IL-2激活后活力增高, 成簇悬浮繁殖、扩增、生长. 在体外, NK细胞可抑制肝癌细胞的增殖、迁移和侵袭. 在体内, NK细胞在裸鼠肝脏可长期存活; NK细胞可明显抑制裸鼠肝癌肺转移. 然而, NK细胞对肝脏肿瘤生长抑制不明显. IL-2可诱导NK细胞免疫相关分子的表达并提高其肿瘤抑制功能.
结论 NK细胞的免疫学功能可被IL-2活化从而抑制肝癌的转移.
Collapse
|
19
|
Ciaglia E, Malfitano AM, Laezza C, Fontana A, Nuzzo G, Cutignano A, Abate M, Pelin M, Sosa S, Bifulco M, Gazzerro P. Immuno-Modulatory and Anti-Inflammatory Effects of Dihydrogracilin A, a Terpene Derived from the Marine Sponge Dendrilla membranosa. Int J Mol Sci 2017; 18:ijms18081643. [PMID: 28788056 PMCID: PMC5578033 DOI: 10.3390/ijms18081643] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/11/2017] [Accepted: 06/23/2017] [Indexed: 01/17/2023] Open
Abstract
We assessed the immunomodulatory and anti-inflammatory effects of 9,11-dihydrogracilin A (DHG), a molecule derived from the Antarctic marine sponge Dendrilla membranosa. We used in vitro and in vivo approaches to establish DHG properties. Human peripheral blood mononuclear cells (PBMC) and human keratinocytes cell line (HaCaT cells) were used as in vitro system, whereas a model of murine cutaneous irritation was adopted for in vivo studies. We observed that DHG reduces dose dependently the proliferative response and viability of mitogen stimulated PBMC. In addition, DHG induces apoptosis as revealed by AnnexinV staining and downregulates the phosphorylation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), signal transducer and activator of transcription (STAT) and extracellular signal–regulated kinase (ERK) at late time points. These effects were accompanied by down-regulation of interleukin 6 (IL-6) production, slight decrease of IL-10 and no inhibition of tumor necrosis factor-alpha (TNF-α) secretion. To assess potential properties of DHG in epidermal inflammation we used HaCaT cells; this compound reduces cell growth, viability and migration. Finally, we adopted for the in vivo study the croton oil-induced ear dermatitis murine model of inflammation. Of note, topical use of DHG significantly decreased mouse ear edema. These results suggest that DHG exerts anti-inflammatory effects and its anti-edema activity in vivo strongly supports its potential therapeutic application in inflammatory cutaneous diseases.
Collapse
Affiliation(s)
- Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081 Baronissi Salerno, Italy.
| | - Anna Maria Malfitano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Salerno, Italy.
| | - Chiara Laezza
- Department of Biology and Cellular and Molecular Pathology, University of Naples Federico II, Via Pansini, 80131 Naples, Italy.
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, Via Pansini 5, 80131 Naples, Italy.
| | - Angelo Fontana
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, Pozzuoli, 80131 Naples; Italy, (A.F.).
| | - Genoveffa Nuzzo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, Pozzuoli, 80131 Naples; Italy, (A.F.).
| | - Adele Cutignano
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, Pozzuoli, 80131 Naples; Italy, (A.F.).
| | - Mario Abate
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081 Baronissi Salerno, Italy.
| | - Marco Pelin
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy.
| | - Silvio Sosa
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy.
| | - Maurizio Bifulco
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Salvatore Allende, 84081 Baronissi Salerno, Italy.
- CORPOREA-Fondazione Idis-Città della Scienza, via Coroglio 104 e 57, 80124 Naples, Italy.
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Salerno, Italy.
| |
Collapse
|
20
|
Li M, Qi Y, Wei J, Lu L, Zhao X, Zhou L. N6-Isopentenyladenosine promoted HeLa cell apoptosis through inhibitions of AKT and transforming growth factor β-activated kinase 1 activation. Tumour Biol 2017; 39:1010428317695966. [PMID: 28345459 DOI: 10.1177/1010428317695966] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
N6-Isopentenyladenosine, a member of the family of plant hormones, possesses anti-cancer activities on a number of cancer cell lines. However, its mode of action in cervical cancer cell remains poorly understood. Our computational docking studies showed that N6-Isopentenyladenosine could bind with the really interesting new gene domain of tumor necrosis factor receptor-associated factor 6, which is an ubiquitination E3 ligase. Tumor necrosis factor receptor-associated factor 6-mediated ubiquitination is known to activate both protein kinase B (also known as AKT) and transforming growth factor β-activated kinase 1, and the really interesting new gene domain comprises the core of the ubiquitin ligase catalytic domain. First, we evaluated the effects of iPA on cervical cancer cell line HeLa using MTT and flow cytometry. Second, we examined the effects of iPA on activation of tumor necrosis factor receptor-associated factor 6-mediated downstream targets using western blot or immunoprecipitation. iPA could reduce HeLa cell proliferation through apoptosis, and such anti-cancer activity is associated with inhibitions of both AKT and transforming growth factor β-activated kinase 1 signaling pathways. In addition, suppression of the anti-apoptotic protein Bcl-2 and elevation of the pro-apoptotic protein Bax were also observed. Anti-proliferation properties of iPA are likely due to its binding at the really interesting new gene domain of tumor necrosis factor receptor-associated factor 6 and loss of AKT and transforming growth factor β-activated kinase 1 activities as a result of functional modulations of tumor necrosis factor receptor-associated factor 6. These results support the emerging notion that tumor necrosis factor receptor-associated factor 6 could serve as a viable target for developing new cancer therapeutics.
Collapse
Affiliation(s)
- Miao Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Yonghao Qi
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Jing Wei
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Lulu Lu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Xuan Zhao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Lijun Zhou
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| |
Collapse
|
21
|
Pisanti S, Malfitano AM, Ciaglia E, Lamberti A, Ranieri R, Cuomo G, Abate M, Faggiana G, Proto MC, Fiore D, Laezza C, Bifulco M. Cannabidiol: State of the art and new challenges for therapeutic applications. Pharmacol Ther 2017; 175:133-150. [PMID: 28232276 DOI: 10.1016/j.pharmthera.2017.02.041] [Citation(s) in RCA: 381] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Over the past years, several lines of evidence support a therapeutic potential of Cannabis derivatives and in particular phytocannabinoids. Δ9-THC and cannabidiol (CBD) are the most abundant phytocannabinoids in Cannabis plants and therapeutic application for both compounds have been suggested. However, CBD is recently emerging as a therapeutic agent in numerous pathological conditions since devoid of the psychoactive side effects exhibited instead by Δ9-THC. In this review, we highlight the pharmacological activities of CBD, its cannabinoid receptor-dependent and -independent action, its biological effects focusing on immunomodulation, angiogenetic properties, and modulation of neuronal and cardiovascular function. Furthermore, the therapeutic potential of cannabidiol is also highlighted, in particular in nuerological diseases and cancer.
Collapse
Affiliation(s)
- Simona Pisanti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Italy.
| | - Anna Maria Malfitano
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Italy
| | - Anna Lamberti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Italy
| | - Roberta Ranieri
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Italy
| | - Gaia Cuomo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Italy
| | - Mario Abate
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Italy
| | - Giorgio Faggiana
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Italy
| | | | | | | | - Maurizio Bifulco
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Italy; Corporea, Fondazione Idis-Città della Scienza, Naples, Italy.
| |
Collapse
|
22
|
Voller J, Maková B, Kadlecová A, Gonzalez G, Strnad M. Plant Hormone Cytokinins for Modulating Human Aging and Age-Related Diseases. HEALTHY AGEING AND LONGEVITY 2017. [DOI: 10.1007/978-3-319-63001-4_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
23
|
Ciaglia E, Abate M, Laezza C, Pisanti S, Vitale M, Seneca V, Torelli G, Franceschelli S, Catapano G, Gazzerro P, Bifulco M. Antiglioma effects of N6-isopentenyladenosine, an endogenous isoprenoid end product, through the downregulation of epidermal growth factor receptor. Int J Cancer 2016; 140:959-972. [PMID: 27813087 DOI: 10.1002/ijc.30505] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 10/25/2016] [Indexed: 12/15/2022]
Abstract
Malignant gliomas are highly dependent on the isoprenoid pathway for the synthesis of lipid moieties critical for cell proliferation. The isoprenoid derivative N6-isopentenyladenosine (iPA) displays pleiotropic biological effects, including a direct anti-tumor activity in several tumor models. The antiglioma effects of iPA was then explored in U87MG cells both in vitro and grafted in mice and the related molecular mechanism confirmed in primary derived patients' glioma cells. iPA powerfully inhibited tumor cell growth and induced caspase-dependent apoptosis through a mechanism involving a marked accumulation of the pro-apoptotic BIM protein and inhibition of EGFR. Indeed, activating AMPK following conversion into its iPAMP active form, iPA stimulated EGFR phosphorylation and ubiquitination along a proteasome-mediated pathway which was responsible for receptor degradation and its downstream signaling pathways inhibition, including the STAT3, ERK and AKT cascade. The inhibition of AMPK by compound C prevented iPA-mediated phosphorylation of EGFR, known to precede receptor loss. As expected the block of EGFR degradation, by exposure to the proteasome inhibitor MG132, significantly reduced iPA-induced cell death. Given the importance of receptor degradation in iPA-mediated cytotoxicity, we also documented that the EGFR expression levels in a panel of primary glioma cells confers them a high sensitivity to iPA treatment. In conclusion our study provides the first evidence of iPA antiglioma effect. Indeed, as glioma is driven by aberrant signaling of growth factor receptors, particularly the EGFR, iPA, alone or in association with EGFR targeted therapies, might be a promising therapeutic tool to achieve a potent anti-tumoral effect.
Collapse
Affiliation(s)
- Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Mario Abate
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, Naples, Italy.,Department of Biology and Cellular and Molecular Pathology, University of Naples Federico II, Naples, Italy
| | - Simona Pisanti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Mario Vitale
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Vincenzo Seneca
- Department of Neurosurgery, "G.Rummo" Medical Hospital, Benevento, Italy
| | - Giovanni Torelli
- Department of Neurosurgery, "San Giovanni di Dio e Ruggi d'Aragona University Hospital", Salerno's School of Medicine, Salerno, Italy
| | | | - Giuseppe Catapano
- Department of Neurosurgery, "G.Rummo" Medical Hospital, Benevento, Italy
| | | | - Maurizio Bifulco
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| |
Collapse
|
24
|
Ciaglia E, Torelli G, Pisanti S, Picardi P, D'Alessandro A, Laezza C, Malfitano AM, Fiore D, Pagano Zottola AC, Proto MC, Catapano G, Gazzerro P, Bifulco M. Cannabinoid receptor CB1 regulates STAT3 activity and its expression dictates the responsiveness to SR141716 treatment in human glioma patients' cells. Oncotarget 2016; 6:15464-81. [PMID: 26008966 PMCID: PMC4558164 DOI: 10.18632/oncotarget.3895] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/29/2015] [Indexed: 12/31/2022] Open
Abstract
Herein we show that a majority of human brain tumor samples and cell lines over-expressed cannabinoid receptor CB1 as compared to normal human astrocytes (NHA), while uniformly expressed low levels of CB2. This finding prompted us to investigate the therapeutic exploitation of CB1 inactivation by SR141716 treatment, with regard to its direct and indirect cell-mediated effects against gliomas. Functional studies, using U251MG glioma cells and primary tumor cell lines derived from glioma patients expressing different levels of CB1, highlighted SR141716 efficacy in inducing apoptosis via G1 phase stasis and block of TGF-β1 secretion through a mechanism that involves STAT3 inhibition. According to the multivariate role of STAT3 in the immune escape too, interestingly SR141716 lead also to the functional and selective expression of MICA/B on the surface of responsive malignant glioma cells, but not on NHA. This makes SR141716 treated-glioma cells potent targets for allogeneic NK cell-mediated recognition through a NKG2D restricted mechanism, thus priming them for NK cell antitumor reactivity. These results indicate that CB1 and STAT3 participate in a new oncogenic network in the complex biology of glioma and their expression levels in patients dictate the efficacy of the CB1 antagonist SR141716 in multimodal glioma destruction.
Collapse
Affiliation(s)
- Elena Ciaglia
- Department of Medicine and Surgery, University of Salerno, Baronissi, Salerno, Italy.,Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Giovanni Torelli
- "G.Rummo" Medical Hospital, Department of Neurosurgery, Benevento, Italy.,Neurosurgery Unit A.O. San Giovanni di Dio e Ruggi d' Aragona - Salerno's School of Medicine, Largo Città di Ippocrate, Salerno, Italy
| | - Simona Pisanti
- Department of Medicine and Surgery, University of Salerno, Baronissi, Salerno, Italy.,Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Paola Picardi
- Department of Medicine and Surgery, University of Salerno, Baronissi, Salerno, Italy.,Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Alba D'Alessandro
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, Naples, Italy.,Department of Biology and Cellular and Molecular Pathology, University of Naples Federico II, Naples, Italy
| | - Anna Maria Malfitano
- Department of Medicine and Surgery, University of Salerno, Baronissi, Salerno, Italy.,Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Donatella Fiore
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | | | | | - Giuseppe Catapano
- "G.Rummo" Medical Hospital, Department of Neurosurgery, Benevento, Italy
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Maurizio Bifulco
- Department of Medicine and Surgery, University of Salerno, Baronissi, Salerno, Italy.,Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| |
Collapse
|
25
|
Hong ZF, Zhao WX, Yin ZY, Xie CR, Xu YP, Chi XQ, Zhang S, Wang XM. Natural killer cells inhibit pulmonary metastasis of hepatocellular carcinoma in nude mice. Oncol Lett 2016; 11:2019-2026. [PMID: 26998115 PMCID: PMC4774462 DOI: 10.3892/ol.2016.4170] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 12/08/2015] [Indexed: 12/29/2022] Open
Abstract
Natural killer (NK) cells have been demonstrated to inhibit tumor growth. However, the role of NK cells in the inhibition of hepatocellular carcinoma metastasis is not well understood. The present study aimed to investigate the roles that NK cells may serve in inhibiting hepatocellular carcinoma metastasis. The role of isolated NK cells in the inhibition, proliferation, migration and invasion of the hepatoma cell line, MHCC97-H, was examined in vitro. Additionally, the survival rate of NK cells labeled with carboxyfluorescein diacetate-succinimidyl ester was assessed in vivo. An orthotopic implantation model was used to evaluate the role of NK cells in suppressing MHCC97-H cells in vivo. The effect of interleukin (IL)-2 stimulation on the tumor-inhibitory role of the NK cells was measured indirectly by analyzing the expression of various NK cell receptors and activated NK cell markers. It was observed that the NK cells inhibited the proliferation, migration and invasion of the MHCC97-H cells in vitro. Furthermore, the NK cells demonstrated long-term survival in the livers of the nude mice, and inhibited lung metastasis of hepatocellular carcinoma in vivo. However, liver tumor growth was not inhibited by the NK cells. IL-2 was identified to enhance the tumor-inhibitory effect of NK cells. The present study concludes that IL-2 may enhance the antitumor activity of the NK cells, and thereby inhibit the metastases of hepatocellular carcinoma in mice.
Collapse
Affiliation(s)
- Zai-Fa Hong
- Department of Hepatobiliary Surgery and Liver Disease Center, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian 361000, P.R. China; Post Graduate College, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Wen-Xiu Zhao
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Zhen-Yu Yin
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Cheng-Rong Xie
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Ya-Ping Xu
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Xiao-Qin Chi
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Sheng Zhang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Xiao-Min Wang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| |
Collapse
|