1
|
Guareschi F, Fonseca C, Silva S, Pescina S, Nicoli S, Buttini F, Sonvico F, Fortuna A. Therapeutic effect of cyclosporine A-loading TPGS micelles on a mouse model of LPS-induced neuroinflammation. Eur J Pharm Sci 2025; 205:106994. [PMID: 39701548 DOI: 10.1016/j.ejps.2024.106994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
Neuroinflammation is an undoubted hallmark of neurodegenerative processes characterized by memory impairment, loss of coordination and muscle strength in diseases such as Alzheimer's disease, Parkinson's disease and multiple sclerosis as well as depressive disorders. Cyclosporine A (CSA) has already been identified as a promising neuroprotective peptide, due to its well-known anti-inflammatory properties. Herein, CSA was encapsulated into α-tocopherol polyethylene glycol 1000 succinate (TPGS) micelles and intranasally administered to a lipopolysaccharide (LPS) induced mouse model of neuroinflammation. After the treatment, mice were subjected to behavioral tests to assess cognitive and motor skills, while the biodistribution of CSA in plasma and olfactory bulb was studied by a new HPLC method validated for precision and accuracy. The results highlighted that in comparison to the classic oral CSA suspension, the intranasal (IN) administration showed significatively better safety and efficiency profiles. Notably, IN administration of CSA micelles showed relevant antidepressive effects and a certain ability to revert LPS-induced motor impairment. This work pointed out that the innovative and noninvasive IN administration of TPGS micelles could represent a safe and effective alternative to the classic oral route to deliver CSA at the Central Nervous System level, where its beneficial activity against neuroinflammation can be exploited.
Collapse
Affiliation(s)
- Fabiola Guareschi
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy
| | - Carla Fonseca
- Laboratory of Pharmacology, Department of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal; Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Soraia Silva
- Laboratory of Pharmacology, Department of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Silvia Pescina
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy
| | - Sara Nicoli
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy
| | - Francesca Buttini
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy; University Research Centre for the Innovation of Health Products, Biopharmanet-TEC, University of Parma, Parma, Italy
| | - Fabio Sonvico
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy; University Research Centre for the Innovation of Health Products, Biopharmanet-TEC, University of Parma, Parma, Italy.
| | - Ana Fortuna
- Laboratory of Pharmacology, Department of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
2
|
Hannon SL, Ding X. Assessing cytochrome P450 function using genetically engineered mouse models. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:253-284. [PMID: 35953157 PMCID: PMC10544722 DOI: 10.1016/bs.apha.2022.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The ability to knock out and/or humanize different genes in experimental animals, globally or in cell- and tissue-specific patterns, has revolutionized scientific research in many areas. Genetically engineered mouse models, including knockout models, transgenic models, and humanized models, have played important roles in revealing the in vivo functions of various cytochrome P450 (CYP) enzymes. These functions are very diverse, ranging from the biotransformation of drugs and other xenobiotics, events that often dictate their pharmacokinetic or toxicokinetic properties and the associated therapeutic or adverse actions, to the metabolism of endogenous compounds, such as steroid hormones and other bioactive substances, that may determine susceptibility to many diseases, such as cancer and metabolic diseases. In this review, we provide a comprehensive list of Cyp-knockout, human CYP-transgenic, and CYP-humanized mouse models that target genes in the CYP1-4 gene families, and highlight their utility in assessing the in vivo metabolism, bioactivation, and toxicity of various xenobiotic compounds, including therapeutic agents and chemical carcinogens. We aim to showcase the advantages of utilizing these mouse models for in vivo drug metabolism and toxicology studies, and to encourage and facilitate greater utility of engineered mouse models to further improve our knowledge of the in vivo functions of various P450 enzymes, which is integral to our ability to develop safer and more effective therapeutics and to identify individuals predisposed to adverse drug reactions or environmental diseases.
Collapse
Affiliation(s)
- Sarrah L Hannon
- Department of Pharmacology and Toxicology, Ken R. Coit College of Pharmacy, The University of Arizona, Tucson, AZ, United States
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, Ken R. Coit College of Pharmacy, The University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
3
|
Borrajo ML, Alonso MJ. Using nanotechnology to deliver biomolecules from nose to brain - peptides, proteins, monoclonal antibodies and RNA. Drug Deliv Transl Res 2022; 12:862-880. [PMID: 34731414 PMCID: PMC8888512 DOI: 10.1007/s13346-021-01086-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
There is a growing number of biomolecules, including peptides, proteins, monoclonal antibodies and RNA, that could be potentially used for the treatment of central nervous system (CNS) diseases. However, the realization of their potential is being hampered by the extraordinary difficulties these complex biomolecules have to reach the brain in therapeutically meaningful amounts. Nose-to-brain (N-to-B) delivery is now being investigated as a potential option for the direct transport of biomolecules from the nasal cavity to different brain areas. Here, we discuss how different technological approaches enhance this N-to-B transport, with emphasis on those that have shown a potential for clinical translation. We also analyse how the physicochemical properties of nanocarriers and their modification with cell-penetrating peptides (CPPs) and targeting ligands affect their efficacy as N-to-B carriers for biomolecules.
Collapse
Affiliation(s)
- Mireya L Borrajo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, 15782, Santiago de Compostela, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, 15782, Santiago de Compostela, Spain.
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
4
|
Kovalchuk N, Zhang QY, Van Winkle L, Ding X. Contribution of Pulmonary CYP-mediated Bioactivation of Naphthalene to Airway Epithelial Injury in the Lung. Toxicol Sci 2021; 177:334-346. [PMID: 32974682 DOI: 10.1093/toxsci/kfaa114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Previous studies have established that cytochrome P450 enzymes (CYPs) in both liver and lung are capable of bioactivating naphthalene (NA), an omnipresent air pollutant and possible human carcinogen, in vitro and in vivo. The aim of this study was to examine the specific contribution of pulmonary CYPs in airway epithelial cells to NA-induced airway toxicity. We used a lung-Cpr-null mouse model, which undergoes doxycycline-induced, Cre-mediated deletion of the Cpr (a redox partner of all microsomal CYPs) gene specifically in airway epithelial cells. In 2-month-old lung-Cpr-null mice, Cpr deletion occurred in 75%-82% of epithelial cells of conducting airways. The extent of NA-induced acute lung toxicity (as indicated by total protein concentration and lactate dehydrogenase activity in bronchoalveolar lavage fluid collected at 24-h after initiation of a 4-h, nose-only, 10-ppm NA inhalation exposure) was substantially lower (by 37%-39%) in lung-Cpr-null mice, compared with control littermates. Moreover, the extent of cellular proliferation (as indicated by 5-bromo-2'-deoxyuridine incorporation) was noticeably lower in both proximal and distal airways (by 59% and 65%, respectively) of NA-treated lung-Cpr-null mice, compared with control littermates, at 2-day post-NA inhalation exposure. A similar genotype-related difference in the extent of postexposure cell proliferation was also observed in mice exposed to NA via intraperitoneal injection at 200 mg/kg. These results directly validate the hypothesis that microsomal CYP enzymes in airway epithelial cells play a large role in causing injury to airway epithelia following exposure to NA via either inhalation or intraperitoneal route.
Collapse
Affiliation(s)
- Nataliia Kovalchuk
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721.,Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York at Albany, Albany, New York 12201
| | - Qing-Yu Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721.,Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York at Albany, Albany, New York 12201
| | - Laura Van Winkle
- Center for Health and the Environment and Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, UC Davis, Davis, California 95616
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
| |
Collapse
|
5
|
Yost EE, Galizia A, Kapraun DF, Persad AS, Vulimiri SV, Angrish M, Lee JS, Druwe IL. Health Effects of Naphthalene Exposure: A Systematic Evidence Map and Analysis of Potential Considerations for Dose-Response Evaluation. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:76002. [PMID: 34251878 PMCID: PMC8274693 DOI: 10.1289/ehp7381] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 05/19/2023]
Abstract
BACKGROUND Naphthalene is a polycyclic aromatic hydrocarbon that has been associated with health effects, including cancer. As the state of the science on naphthalene toxicity continues to evolve, updated toxicity reference value(s) may be required to support human health risk assessment. OBJECTIVES We present a systematic evidence map of studies that could be used to derive toxicity reference value(s) for naphthalene. METHODS Human and animal health effect studies and physiologically based pharmacokinetic (PBPK) models were identified from a literature search based on populations, exposures, comparators, and outcomes (PECO) criteria. Human and animal studies meeting PECO criteria were refined to a smaller subset considered most informative for deriving chronic reference value(s), which are preferred for assessing risk to the general public. This subset was evaluated for risk of bias and sensitivity, and the suitability of each study for dose-response analysis was qualitatively assessed. Lowest observed adverse effect levels (LOAELs) were extracted and summarized. Other potentially relevant studies (e.g., mechanistic and toxicokinetic studies) were tracked as supplemental information but not evaluated further. Existing reference values for naphthalene are also summarized. RESULTS We identified 26 epidemiology studies and 16 animal studies that were considered most informative for further analysis. Eleven PBPK models were identified. The available epidemiology studies generally had significant risk of bias and/or sensitivity concerns and were mostly found to have low suitability for dose-response analysis due to the nature of the exposure measurements. The animal studies had fewer risk of bias and sensitivity concerns and were mostly found to be suitable for dose-response analysis. CONCLUSION Although both epidemiological and animal studies of naphthalene provide weight of evidence for hazard identification, the available animal studies appear more suitable for reference value derivation. PBPK models and mechanistic and toxicokinetic data can be applied to extrapolate these animal data to humans, considering mode of action and interspecies metabolic differences. https://doi.org/10.1289/EHP7381.
Collapse
Affiliation(s)
- Erin E. Yost
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Audrey Galizia
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Dustin F. Kapraun
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Amanda S. Persad
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Suryanarayana V. Vulimiri
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Michelle Angrish
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Janice S. Lee
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Ingrid L. Druwe
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| |
Collapse
|
6
|
Kovalchuk N, Zhang QY, Kelty J, Van Winkle L, Ding X. Toxicokinetic Interaction between Hepatic Disposition and Pulmonary Bioactivation of Inhaled Naphthalene Studied Using Cyp2abfgs-Null and CYP2A13/2F1-Humanized Mice with Deficient Hepatic Cytochrome P450 Activity. Drug Metab Dispos 2019; 47:1469-1478. [PMID: 31594800 PMCID: PMC7042723 DOI: 10.1124/dmd.119.088930] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/01/2019] [Indexed: 11/22/2022] Open
Abstract
Previous studies using Cyp2abfgs-null (lacking all genes of the Cyp2a, 2b, 2f, 2g, and 2s subfamilies), CYP2A13/2F1-humanized, and liver-Cpr-null (LCN) mice showed that although hepatic cytochrome P450 (P450) enzymes are essential for systemic clearance of inhaled naphthalene (a possible human carcinogen), both hepatic and extrahepatic P450 enzymes may contribute to naphthalene-induced lung toxicity via bioactivation. Herein, we aimed to further understand the toxicokinetics of inhaled naphthalene in order to provide a basis for predicting the effects of variations in rates of xenobiotic disposition on the extent of target tissue bioactivation. We assessed the impact of a hepatic deficit in naphthalene metabolism on the toxicokinetics of inhaled naphthalene using newly generated Cyp2abfgs-null-and-LCN and CYP2A13/2F1-humanized-and-LCN mice. We determined plasma, lung, and liver levels of naphthalene and naphthalene-glutathione conjugate, a biomarker of naphthalene bioactivation, over time after naphthalene inhalation. We found that the loss of hepatic naphthalene metabolism severely decreased naphthalene systemic clearance and caused naphthalene to accumulate in the liver and other tissues. Naphthalene release from tissue, as evidenced by the continued increase in plasma naphthalene levels after termination of active inhalation exposure, was accompanied by prolonged bioactivation of naphthalene in the lung. In addition, transgenic expression of human CYP2A13/2F1 in the respiratory tract caused a reduction in plasma naphthalene levels (by 40%, relative to Cyp2abfgs-null-and-LCN mice) and corresponding decreases in naphthalene-glutathione levels in the lung in mice with hepatic P450 deficiency, despite the increase in local naphthalene-bioactivating P450 activity. Thus, the bioavailability of naphthalene in the target tissue has a significant effect on the extent of naphthalene bioactivation in the lung. SIGNIFICANCE STATEMENT: In this study, we report several novel findings related to the toxicokinetics of inhaled naphthalene, the ability of which to cause lung carcinogenesis in humans is a current topic for risk assessment. We show the accumulation of naphthalene in the liver and lung in mice with compromised hepatic cytochrome P450 (P450) activity; the ability of tissue-stored naphthalene to redistribute to the circulation after termination of active inhalation exposure, prolonging exposure of target tissues to naphthalene; and the ability of non-CYP2ABFGS enzymes of the lung to bioactivate naphthalene. These results suggest potentially large effects of deficiencies in hepatic P450 activity on naphthalene tissue burden and bioactivation in human lungs.
Collapse
Affiliation(s)
- Nataliia Kovalchuk
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (N.K., Q.-Y.Z., X.D.); Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York (N.K., Q.-Y.Z.); Center for Health and the Environment, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, California (J.K., L.V.W.); and College of Nanoscale Science, State University of New York Polytechnic Institute, Albany, New York (X.D.)
| | - Qing-Yu Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (N.K., Q.-Y.Z., X.D.); Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York (N.K., Q.-Y.Z.); Center for Health and the Environment, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, California (J.K., L.V.W.); and College of Nanoscale Science, State University of New York Polytechnic Institute, Albany, New York (X.D.)
| | - Jacklyn Kelty
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (N.K., Q.-Y.Z., X.D.); Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York (N.K., Q.-Y.Z.); Center for Health and the Environment, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, California (J.K., L.V.W.); and College of Nanoscale Science, State University of New York Polytechnic Institute, Albany, New York (X.D.)
| | - Laura Van Winkle
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (N.K., Q.-Y.Z., X.D.); Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York (N.K., Q.-Y.Z.); Center for Health and the Environment, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, California (J.K., L.V.W.); and College of Nanoscale Science, State University of New York Polytechnic Institute, Albany, New York (X.D.)
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (N.K., Q.-Y.Z., X.D.); Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York (N.K., Q.-Y.Z.); Center for Health and the Environment, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, California (J.K., L.V.W.); and College of Nanoscale Science, State University of New York Polytechnic Institute, Albany, New York (X.D.)
| |
Collapse
|
7
|
Hartog M, Zhang QY, Ding X. Role of mouse cytochrome P450 enzymes of the CYP2ABFGS subfamilies in the induction of lung inflammation by cigarette smoke exposure. Toxicol Sci 2019; 172:123-131. [PMID: 31388674 PMCID: PMC6813748 DOI: 10.1093/toxsci/kfz171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/01/2019] [Accepted: 06/27/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Many constituents of tobacco smoke (TS) require bioactivation to exert toxic effects; however, few studies have examined the role of bioactivation enzymes in the adverse effects of TS exposure. This knowledge gap is a major source of uncertainty for risk assessment and chemoprevention efforts. OBJECTIVES Our aim is to test the hypothesis that cytochrome P450 (P450) enzyme mediated bioactivation is essential to the development of TS exposure-induced lung toxicity, by determining the contributions of P450 enzymes in the mouse Cyp2abfgs gene subfamilies to environmental tobacco smoke (ETS)-induced lung inflammation. METHODS Adult female wildtype (WT) and Cyp2abfgs-null mice (both on C57BL/6J background) were exposed to filtered air or ETS, intermittently, for 1 or 2 weeks. Lung inflammation was assessed by quantification of inflammatory cells, cytokines, chemokines, proteins in bronchoalveolar lavage fluid (BALF), and histopathological analysis. Glutathione (GSH) conjugates of two ETS constituents, naphthalene (NA) and 3-methylindole (3MI), were measured in mice exposed to ETS for four hours. RESULTS Persistent macrophagic and neutrophilic lung inflammation was observed in ETS-exposed WT mice; the extent of which was significantly reduced in ETS-exposed Cyp2abfgs-null mice. Levels of proinflammatory cytokines and chemokines, along with the total protein concentration, were increased in cell-free BALF from ETS-exposed WT mice, but not Cyp2abfgs-null mice. Additionally, GSH-conjugates of NA and 3MI were detected in the lungs of WT, but not Cyp2abfgs-null, mice following ETS exposure. CONCLUSIONS These results provide the first in vivo evidence that the mouse Cyp2abfgs gene cluster plays an important role in ETS-induced lung inflammation.
Collapse
Affiliation(s)
- Matthew Hartog
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY
| | - Qing-Yu Zhang
- Wadsworth Center, New York State Department of Health, Albany, NY.,Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson
| | - Xinxin Ding
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY.,Wadsworth Center, New York State Department of Health, Albany, NY.,Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson
| |
Collapse
|
8
|
Heydel JM, Faure P, Neiers F. Nasal odorant metabolism: enzymes, activity and function in olfaction. Drug Metab Rev 2019; 51:224-245. [DOI: 10.1080/03602532.2019.1632890] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jean-Marie Heydel
- Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Philippe Faure
- Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Fabrice Neiers
- Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| |
Collapse
|
9
|
Torres-Ortega PV, Saludas L, Hanafy AS, Garbayo E, Blanco-Prieto MJ. Micro- and nanotechnology approaches to improve Parkinson's disease therapy. J Control Release 2018; 295:201-213. [PMID: 30579984 DOI: 10.1016/j.jconrel.2018.12.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022]
Abstract
Current therapies for Parkinson's disease are symptomatic and unable to regenerate the brain tissue. In recent years, the therapeutic potential of a wide variety of neuroprotective and neuroregenerative molecules such as neurotrophic factors, antioxidants and RNA-based therapeutics has been explored. However, drug delivery to the brain is still a challenge and the therapeutic efficacy of many drugs is limited. In the last decade, micro- and nanoparticles have proved to be powerful tools for the administration of these molecules to the brain, enabling the development of new strategies against Parkinson's disease. The list of encapsulated drugs and the nature of the particles used is long, and numerous studies have been carried out supporting their efficacy in treating this pathology. This review aims to give an overview of the latest advances and emerging frontiers in micro- and nanomedical approaches for repairing dopaminergic neurons. Special emphasis will be placed on offering a new perspective to link these advances with the most relevant clinical trials and with the real possibility of transferring micro- and nanoformulations to industrial scale-up processes. This review is intended as a contribution towards facing the challenges that still exist in the clinical translation of micro- and nanotechnologies to administer therapeutic agents in Parkinson's disease.
Collapse
Affiliation(s)
- Pablo Vicente Torres-Ortega
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Laura Saludas
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Amira Sayed Hanafy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria (PUA), Alexandria, Egypt
| | - Elisa Garbayo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain.
| | - María José Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain.
| |
Collapse
|
10
|
Fu Z, Ogura T, Luo W, Lin W. ATP and Odor Mixture Activate TRPM5-Expressing Microvillous Cells and Potentially Induce Acetylcholine Release to Enhance Supporting Cell Endocytosis in Mouse Main Olfactory Epithelium. Front Cell Neurosci 2018; 12:71. [PMID: 29615870 PMCID: PMC5869921 DOI: 10.3389/fncel.2018.00071] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/27/2018] [Indexed: 12/13/2022] Open
Abstract
The main olfactory epithelium (MOE) functions to detect odor molecules, provide an epithelial surface barrier, and remove xenobiotics from inhaled air. Mechanisms coordinating the activities of different cell types within the MOE to maintain these functions are poorly understood. Previously, we showed that superficially located microvillous cells (MCs) in the MOE expressing transient receptor potential channel M5 (TRPM5) are cholinergic and chemoresponsive and that they play an important role in maintaining odor responses and olfactory-guided behavior under challenging chemical environment. Here we investigated TRPM5-MC activation and subsequent paracrine regulation. Ca2+ imaging showed that TRPM5-MCs dose-dependently increase their intracellular Ca2+ levels in response to ATP, an important signaling molecule for airway mucociliary movement, and to an odor mixture. Pharmacological examination showed that the ATP responses are primarily mediated by P2X purinergic receptors. Interestingly, using the endocytosis dye pHrodo Red dextran, we found that chemical-activated TRPM5-MCs significantly increase the number of pHrodo-labeled puncta compared to controls without stimulation and compared to cells that do not respond to ATP or to the odor mixture. These results indicate potential vesicle recycling after release of the signaling molecule acetylcholine (ACh). Interestingly, TRPM5 knockout (KO) results in a decrease in ATP-induced pHrodo internalization. We further investigated cholinergic regulation of neighboring supporting cells (SCs). We found that ACh strongly elevates intracellular Ca2+ and potentiates pHrodo endocytosis in SCs. The ACh effects are diminished in the presence of atropine or M3 muscarinic receptor antagonist and in SCs lacking M3 receptors. Collectively, these data suggest that TRPM5-MCs may regulate the MOE’s multicellular network activity via cholinergic paracrine signaling for functional maintenance and adaptive plasticity.
Collapse
Affiliation(s)
- Ziying Fu
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD, United States
| | - Tatsuya Ogura
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD, United States
| | - Wangmei Luo
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD, United States
| | - Weihong Lin
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD, United States
| |
Collapse
|
11
|
Samaridou E, Alonso MJ. Nose-to-brain peptide delivery - The potential of nanotechnology. Bioorg Med Chem 2017; 26:2888-2905. [PMID: 29170026 DOI: 10.1016/j.bmc.2017.11.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/26/2017] [Accepted: 11/02/2017] [Indexed: 12/11/2022]
Abstract
Nose-to-brain (N-to-B) delivery offers to protein and peptide drugs the possibility to reach the brain in a non-invasive way. This article is a comprehensive review of the state-of-the-art of this emerging peptide delivery route, as well as of the challenges associated to it. Emphasis is given on the potential of nanosized drug delivery carriers to enhance the direct N-to-B transport of protein or peptide drugs. In particular, polymer- and lipid- based nanocarriers are comparatively analyzed in terms of the influence of their physicochemical characteristics and composition on their in vivo fate and efficacy. The use of biorecognitive ligands and permeation enhancers in order to enhance their brain targeting efficiency is also discussed. The article concludes highlighting the early stage of this research field and its still unveiled potential. The final message is that more explicatory PK/PD studies are required in order to achieve the translation from preclinical to the clinical development phase.
Collapse
Affiliation(s)
- Eleni Samaridou
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Av. Barcelona s/n, Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Maria José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Av. Barcelona s/n, Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
12
|
Li L, Carratt S, Hartog M, Kovalchik N, Jia K, Wang Y, Zhang QY, Edwards P, Winkle LV, Ding X. Human CYP2A13 and CYP2F1 Mediate Naphthalene Toxicity in the Lung and Nasal Mucosa of CYP2A13/2F1-Humanized Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:067004. [PMID: 28599267 PMCID: PMC5743450 DOI: 10.1289/ehp844] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/22/2016] [Accepted: 11/07/2016] [Indexed: 05/29/2023]
Abstract
BACKGROUND The potential carcinogenicity of naphthalene (NA), a ubiquitous environmental pollutant, in human respiratory tract is a subject of intense debate. Chief among the uncertainties in risk assessment for NA is whether human lung CYP2A13 and CYP2F1 can mediate NA's respiratory tract toxicity. OBJECTIVES We aimed to assess the in vivo function of CYP2A13 and CYP2F1 in NA bioactivation and NA-induced respiratory tract toxicity in mouse models. METHODS Rates of microsomal NA bioactivation and the effects of an anti-CYP2A antibody were determined for lung and nasal olfactory mucosa (OM) from Cyp2abfgs-null, CYP2A13-humanized, and CYP2A13/2F1-humanized mice. The extent of NA respiratory toxicity was compared among wild-type, Cyp2abfgs-null, and CYP2A13/2F1-humanized mice following inhalation exposure at an occupationally relevant dose (10 ppm for 4 hr). RESULTS In vitro studies indicated that the NA bioactivation activities in OM and lung of the CYP2A13/2F1-humanized mice were primarily contributed by, respectively, CYP2A13 and CYP2F1. CYP2A13/2F1-humanized mice showed greater sensitivity to NA than Cyp2abfgs-null mice, with greater depletion of nonprotein sulfhydryl and occurrence of cytotoxicity (observable by routine histology) in the OM, at 2 or 20 hr after termination of NA exposure, in humanized mice. Focal, rather than gross, lung toxicity was observed in Cyp2abfgs-null and CYP2A13/2F1-humanized mice; however, the extent of NA-induced lung injury (shown as volume fraction of damaged cells) was significantly greater in the terminal bronchioles of CYP2A13/2F1-humanized mice than in Cyp2abfgs-null mice. CONCLUSION CYP2F1 is an active enzyme. Both CYP2A13 and CYP2F1 are active toward NA in the CYP2A13/2F1-humanized mice, where they play significant roles in NA-induced respiratory tract toxicity. https://doi.org/10.1289/EHP844.
Collapse
Affiliation(s)
- Lei Li
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Sarah Carratt
- Center for Health and the Environment, University of California, Davis (UC Davis), Davis, California, USA
| | - Matthew Hartog
- College of Nanoscale Science and Engineering, State University of New York (SUNY) Polytechnic Institute, Albany, New York, USA
| | - Nataliia Kovalchik
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Kunzhi Jia
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Yanan Wang
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Qing-Yu Zhang
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Patricia Edwards
- Center for Health and the Environment, University of California, Davis (UC Davis), Davis, California, USA
| | - Laura Van Winkle
- Center for Health and the Environment, University of California, Davis (UC Davis), Davis, California, USA
| | - Xinxin Ding
- College of Nanoscale Science and Engineering, State University of New York (SUNY) Polytechnic Institute, Albany, New York, USA
| |
Collapse
|
13
|
Impact of hepatic P450-mediated biotransformation on the disposition and respiratory tract toxicity of inhaled naphthalene. Toxicol Appl Pharmacol 2017; 329:1-8. [PMID: 28527914 DOI: 10.1016/j.taap.2017.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/14/2017] [Accepted: 05/15/2017] [Indexed: 11/23/2022]
Abstract
We determined whether a decrease in hepatic microsomal cytochrome P450 activity would impact lung toxicity induced by inhalation exposure to naphthalene (NA), a ubiquitous environmental pollutant. The liver-Cpr-null (LCN) mouse showed decreases in microsomal metabolism of NA in liver, but not lung, compared to wild-type (WT) mouse. Plasma levels of NA and NA-glutathione conjugates (NA-GSH) were both higher in LCN than in WT mice after a 4-h nose-only NA inhalation exposure at 10ppm. Levels of NA were also higher in lung and liver of LCN, compared to WT, mice, following exposure to NA at 5 or 10ppm. Despite the large increase in circulating and lung tissue NA levels, the level of NA-GSH, a biomarker of NA bioactivation, was either not different, or only slightly higher, in lung and liver tissues of LCN mice, relative to that in WT mice. Furthermore, the extent of NA-induced acute airway injury, judging from high-resolution lung histopathology and morphometry at 20h following NA exposure, was not higher, but lower, in LCN than in WT mice. These results, while confirming the ability of extrahepatic organ to bioactivate inhaled NA and mediate NA's lung toxicity, suggest that liver P450-generated NA metabolites also have a significant, although relatively small, contribution to airway toxicity of inhaled NA. This hepatic contribution to the airway toxicity of inhaled NA may be an important risk factor for individuals with diminished bioactivation activity in the lung.
Collapse
|
14
|
Shimada T, Takenaka S, Kakimoto K, Murayama N, Lim YR, Kim D, Foroozesh MK, Yamazaki H, Guengerich FP, Komori M. Structure-Function Studies of Naphthalene, Phenanthrene, Biphenyl, and Their Derivatives in Interaction with and Oxidation by Cytochromes P450 2A13 and 2A6. Chem Res Toxicol 2016; 29:1029-40. [PMID: 27137136 PMCID: PMC5293596 DOI: 10.1021/acs.chemrestox.6b00083] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Naphthalene, phenanthrene, biphenyl, and their derivatives having different ethynyl, propynyl, butynyl, and propargyl ether substitutions were examined for their interaction with and oxidation by cytochromes P450 (P450) 2A13 and 2A6. Spectral interaction studies suggested that most of these chemicals interacted with P450 2A13 to induce Type I binding spectra more readily than with P450 2A6. Among the various substituted derivatives examined, 2-ethynylnaphthalene, 2-naphthalene propargyl ether, 3-ethynylphenanthrene, and 4-biphenyl propargyl ether had larger ΔAmax/Ks values in inducing Type I binding spectra with P450 2A13 than their parent compounds. P450 2A13 was found to oxidize naphthalene, phenanthrene, and biphenyl to 1-naphthol, 9-hydroxyphenanthrene, and 2- and/or 4-hydroxybiphenyl, respectively, at much higher rates than P450 2A6. Other human P450 enzymes including P450s 1A1, 1A2, 1B1, 2C9, and 3A4 had lower rates of oxidation of naphthalene, phenanthrene, and biphenyl than P450s 2A13 and 2A6. Those alkynylated derivatives that strongly induced Type I binding spectra with P450s 2A13 and 2A6 were extensively oxidized by these enzymes upon analysis with HPLC. Molecular docking studies supported the hypothesis that ligand-interaction energies (U values) obtained with reported crystal structures of P450 2A13 and 2A6 bound to 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, indole, pilocarpine, nicotine, and coumarin are of use in understanding the basis of possible molecular interactions of these xenobiotic chemicals with the active sites of P450 2A13 and 2A6 enzymes. In fact, the ligand-interaction energies with P450 2A13 4EJG bound to these chemicals were found to relate to their induction of Type I binding spectra.
Collapse
Affiliation(s)
- Tsutomu Shimada
- Laboratory of Cellular and Molecular Biology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Shigeo Takenaka
- Laboratory of Cellular and Molecular Biology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Kensaku Kakimoto
- Department of Biological Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Norie Murayama
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Young-Ran Lim
- Department of Biological Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Donghak Kim
- Department of Biological Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Maryam K. Foroozesh
- Department of Chemistry, Xavier University of Louisiana, New Orleans, Louisiana 70125, United States
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - F. Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Masayuki Komori
- Laboratory of Cellular and Molecular Biology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
15
|
Bailey LA, Nascarella MA, Kerper LE, Rhomberg LR. Hypothesis-based weight-of-evidence evaluation and risk assessment for naphthalene carcinogenesis. Crit Rev Toxicol 2015; 46:1-42. [PMID: 26202831 PMCID: PMC4732411 DOI: 10.3109/10408444.2015.1061477] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/09/2015] [Indexed: 11/13/2022]
Abstract
Inhalation of naphthalene causes olfactory epithelial nasal tumors in rats (but not in mice) and benign lung adenomas in mice (but not in rats). The limited available human data have not identified an association between naphthalene exposure and increased respiratory cancer risk. Assessing naphthalene's carcinogenicity in humans, therefore, depends entirely on experimental evidence from rodents. We evaluated the respiratory carcinogenicity of naphthalene in rodents, and its potential relevance to humans, using our Hypothesis-Based Weight-of-Evidence (HBWoE) approach. We systematically and comparatively reviewed data relevant to key elements in the hypothesized modes of action (MoA) to determine which is best supported by the available data, allowing all of the data from each realm of investigation to inform interpretation of one another. Our analysis supports a mechanism that involves initial metabolism of naphthalene to the epoxide, followed by GSH depletion, cytotoxicity, chronic inflammation, regenerative hyperplasia, and tumor formation, with possible weak genotoxicity from downstream metabolites occurring only at high cytotoxic doses, strongly supporting a non-mutagenic threshold MoA in the rat nose. We also conducted a dose-response analysis, based on the likely MoA, which suggests that the rat nasal MoA is not relevant in human respiratory tissues at typical environmental exposures. Our analysis illustrates how a thorough WoE evaluation can be used to support a MoA, even when a mechanism of action cannot be fully elucidated. A non-mutagenic threshold MoA for naphthalene-induced rat nasal tumors should be considered as a basis to determine human relevance and to guide regulatory and risk-management decisions.
Collapse
|