51
|
Hilliard KL, Allen E, Traber KE, Yamamoto K, Stauffer NM, Wasserman GA, Jones MR, Mizgerd JP, Quinton LJ. The Lung-Liver Axis: A Requirement for Maximal Innate Immunity and Hepatoprotection during Pneumonia. Am J Respir Cell Mol Biol 2015; 53:378-90. [PMID: 25607543 DOI: 10.1165/rcmb.2014-0195oc] [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] [Indexed: 12/21/2022] Open
Abstract
The hepatic acute-phase response (APR), stimulated by injury or inflammation, is characterized by significant changes in circulating acute-phase protein (APP) concentrations. Although individual functions of liver-derived APPs are known, the net consequence of APP changes is unclear. Pneumonia, which induces the APR, causes an inflammatory response within the airspaces that is coordinated largely by alveolar macrophages and is typified by cytokine production, leukocyte recruitment, and plasma extravasation, the latter of which may enable delivery of hepatocyte-derived APPs to the infection site. To determine the functional significance of the hepatic APR during pneumonia, we challenged APR-null mice lacking hepatocyte signal transducer and activator of transcription 3 (STAT3) and v-rel avian reticuloendotheliosis viral oncogene homolog A (RelA) with Escherichia coli in the airspaces. APR-null mice displayed ablated APP induction, significantly increased mortality, liver injury and apoptosis, and a trend toward increased bacterial burdens. TNF-α neutralization reversed hepatotoxicity, but not mortality, suggesting that APR-dependent survival is not solely due to hepatoprotection. After a milder (nonlethal) E. coli infection, hepatocyte-specific mutations decreased APP concentrations and pulmonary inflammation in bronchoalveolar lavage fluid. Cytokine expression in airspace macrophages, but not other airspace or circulating cells, was significantly dependent on APP extravasation into the alveoli. These data identify a novel signaling axis whereby the liver response enhances macrophage activation and pulmonary inflammation during pneumonia. Although hepatic acute-phase changes directly curb injury induced by TNF-α in the liver itself, APPs downstream of these same signals promote survival in association with innate immunity in the lungs, thus demonstrating a critical role for the lung-liver axis during pneumonia.
Collapse
Affiliation(s)
- Kristie L Hilliard
- Departments of 1 Microbiology.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Eri Allen
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Katrina E Traber
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Kazuko Yamamoto
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Nicole M Stauffer
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Gregory A Wasserman
- Departments of 1 Microbiology.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Matthew R Jones
- 3 Medicine.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Joseph P Mizgerd
- Departments of 1 Microbiology.,3 Medicine.,4 Biochemistry, and.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Lee J Quinton
- 3 Medicine.,5 Pathology and Laboratory Medicine, and.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
52
|
Palaniyandi S, Liu X, Periasamy S, Ma A, Tang J, Jenkins M, Tuo W, Song W, Keegan AD, Conrad DH, Zhu X. Inhibition of CD23-mediated IgE transcytosis suppresses the initiation and development of allergic airway inflammation. Mucosal Immunol 2015; 8:1262-74. [PMID: 25783969 PMCID: PMC4575230 DOI: 10.1038/mi.2015.16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 02/02/2015] [Indexed: 02/04/2023]
Abstract
The epithelial lining of the airway tract and allergen-specific IgE are considered essential controllers of inflammatory responses to allergens. The human low affinity IgE receptor, CD23 (FcɛRII), is capable of transporting IgE or IgE-allergen complexes across the polarized human airway epithelial cell (AEC) monolayer in vitro. However, it remains unknown whether the CD23-dependent IgE transfer pathway in AECs initiates and facilitates allergic inflammation in vivo, and whether inhibition of this pathway attenuates allergic inflammation. To this end, we show that in wild-type (WT) mice, epithelial CD23 transcytosed both IgE and ovalbumin (OVA)-IgE complexes across the airway epithelial barrier, whereas neither type of transcytosis was observed in CD23 knockout (KO) mice. In chimeric mice, OVA sensitization and aerosol challenge of WT/WT (bone-marrow transfer from the WT to WT) or CD23KO/WT (CD23KO to WT) chimeric mice, which express CD23 on radioresistant airway structural cells (mainly epithelial cells) resulted in airway eosinophilia, including collagen deposition and a significant increase in goblet cells, and increased airway hyperreactivity. In contrast, the absence of CD23 expression on airway structural or epithelial cells, but not on hematopoietic cells, in WT/CD23KO (the WT to CD23KO) chimeric mice significantly reduced OVA-driven allergic airway inflammation. In addition, inhalation of the CD23-blocking B3B4 antibody in sensitized WT mice before or during airway challenge suppressed the salient features of asthma, including bronchial hyperreactivity. Taken together, these results identify a previously unproven mechanism in which epithelial CD23 plays a central role in the development of allergic inflammation. Further, our study suggests that functional inhibition of CD23 in the airway is a potential therapeutic approach to inhibit the development of asthma.
Collapse
Affiliation(s)
- Senthilkumar Palaniyandi
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA,Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA
| | - Xiaoyang Liu
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Sivakumar Periasamy
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Aiying Ma
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | | | - Mark Jenkins
- Animal Parasitic Diseases Laboratory, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Wenbin Tuo
- Animal Parasitic Diseases Laboratory, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Wenxia Song
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA,Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA
| | - Achsah D. Keegan
- Center for Vascular and Inflammatory Diseases and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Daniel H. Conrad
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Xiaoping Zhu
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA,Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA,To whom all correspondence should be addressed: Dr. Xiaoping Zhu, VA-MD Regional College of Veterinary Medicine, University of Maryland, 8075 Greenmead Drive, College Park, MD 20742, USA, Telephone: (301)314-6814; Fax: (301)314-6855,
| |
Collapse
|
53
|
Konduru NV, Jimenez RJ, Swami A, Friend S, Castranova V, Demokritou P, Brain JD, Molina RM. Silica coating influences the corona and biokinetics of cerium oxide nanoparticles. Part Fibre Toxicol 2015; 12:31. [PMID: 26458946 PMCID: PMC4603643 DOI: 10.1186/s12989-015-0106-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/28/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The physicochemical properties of nanoparticles (NPs) influence their biological outcomes. METHODS We assessed the effects of an amorphous silica coating on the pharmacokinetics and pulmonary effects of CeO2 NPs following intratracheal (IT) instillation, gavage and intravenous injection in rats. Uncoated and silica-coated CeO2 NPs were generated by flame spray pyrolysis and later neutron-activated. These radioactive NPs were IT-instilled, gavaged, or intravenously (IV) injected in rats. Animals were analyzed over 28 days post-IT, 7 days post-gavage and 2 days post-injection. RESULTS Our data indicate that silica coating caused more but transient lung inflammation compared to uncoated CeO2. The transient inflammation of silica-coated CeO2 was accompanied by its enhanced clearance. Then, from 7 to 28 days, clearance was similar although significantly more (141)Ce from silica-coated (35%) was cleared than from uncoated (19%) (141)CeO2 in 28 days. The protein coronas of the two NPs were significantly different when they were incubated with alveolar lining fluid. Despite more rapid clearance from the lungs, the extrapulmonary (141)Ce from silica-coated (141)CeO2 was still minimal (<1%) although lower than from uncoated (141)CeO2 NPs. Post-gavage, nearly 100% of both NPs were excreted in the feces consistent with very low gut absorption. Both IV-injected (141)CeO2 NP types were primarily retained in the liver and spleen. The silica coating significantly altered the plasma protein corona composition and enhanced retention of (141)Ce in other organs except the liver. CONCLUSION We conclude that silica coating of nanoceria alters the biodistribution of cerium likely due to modifications in protein corona formation after IT and IV administration.
Collapse
Affiliation(s)
- Nagarjun V Konduru
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Renato J Jimenez
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Archana Swami
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Sherri Friend
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Vincent Castranova
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, P.O. Box 9530, Morgantown, WV, 26506, USA
| | - Philip Demokritou
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Joseph D Brain
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Ramon M Molina
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
54
|
Efficacy of aerosol therapy of lung cancer correlates with EGFR paralysis induced by AvidinOX-anchored biotinylated Cetuximab. Oncotarget 2015; 5:9239-55. [PMID: 25238453 PMCID: PMC4253431 DOI: 10.18632/oncotarget.2409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Lung cancer, as well as lung metastases from distal primary tumors, could benefit from aerosol treatment. Unfortunately, because of lung physiology, clearance of nebulized drugs is fast, paralleled by unwanted systemic exposure. Here we report that nebulized AvidinOX can act as an artificial receptor for biotinylated drugs. In nude and SCID mice with advanced human KRAS-mutated A549 metastatic lung cancer, pre-nebulization with AvidinOX enables biotinylated Cetuximab to control tumor growth at a dose lower than 1/25,000 the intravenous effective dose. This result correlates with a striking, specific and unpredictable effect of AvidinOX-anchored biotinylated Cetuximab, as well as Panitumumab, observed on a panel of tumor cell lines, leading to inhibition of dimerization and signalling, blockade of endocytosis, induction of massive lysosomal degradation and abrogation of nuclear translocation of EGFR. Excellent tolerability, together with availability of pharmaceutical-grade AvidinOX and antibodies, will allow rapid clinical translation of the proposed therapy.
Collapse
|
55
|
Woods A, Patel A, Spina D, Riffo-Vasquez Y, Babin-Morgan A, de Rosales RTM, Sunassee K, Clark S, Collins H, Bruce K, Dailey LA, Forbes B. In vivo biocompatibility, clearance, and biodistribution of albumin vehicles for pulmonary drug delivery. J Control Release 2015; 210:1-9. [PMID: 25980621 PMCID: PMC4674532 DOI: 10.1016/j.jconrel.2015.05.269] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/06/2015] [Accepted: 05/11/2015] [Indexed: 01/31/2023]
Abstract
The development of clinically acceptable albumin-based nanoparticle formulations for use in pulmonary drug delivery has been hindered by concerns about the toxicity of nanomaterials in the lungs combined with a lack of information on albumin nanoparticle clearance kinetics and biodistribution. In this study, the in vivo biocompatibility of albumin nanoparticles was investigated following a single administration of 2, 20, and 390 μg/mouse, showing no inflammatory response (TNF-α and IL-6, cellular infiltration and protein concentration) compared to vehicle controls at the two lower doses, but elevated mononucleocytes and a mild inflammatory effect at the highest dose tested. The biodistribution and clearance of 111In labelled albumin solution and nanoparticles over 48 h following a single pulmonary administration to mice was investigated by single photon emission computed tomography and X-ray computed tomography imaging and terminal biodistribution studies. 111In labelled albumin nanoparticles were cleared more slowly from the mouse lung than 111In albumin solution (64.1 ± 8.5% vs 40.6 ± 3.3% at t = 48 h, respectively), with significantly higher (P < 0.001) levels of albumin nanoparticle-associated radioactivity located within the lung tissue (23.3 ± 4.7%) compared to the lung fluid (16.1 ± 4.4%). Low amounts of 111In activity were detected in the liver, kidneys, and intestine at time points > 24 h indicating that small amounts of activity were cleared from the lungs both by translocation across the lung mucosal barrier, as well as mucociliary clearance. This study provides important information on the fate of albumin vehicles in the lungs, which may be used to direct future formulation design of inhaled nanomedicines.
Collapse
Affiliation(s)
- A Woods
- Drug Delivery Research Group, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - A Patel
- Drug Delivery Research Group, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, United Kingdom; Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - D Spina
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Y Riffo-Vasquez
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - A Babin-Morgan
- Drug Delivery Research Group, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, United Kingdom; Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - R T M de Rosales
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - K Sunassee
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - S Clark
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - H Collins
- Division of Immunology, Infection & Inflammatory Diseases, Guy's Campus, King's College London, 15-16 Newcomen Street, London SE1 1UL, United Kingdom
| | - K Bruce
- Drug Delivery Research Group, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - L A Dailey
- Drug Delivery Research Group, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, United Kingdom.
| | - B Forbes
- Drug Delivery Research Group, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, United Kingdom
| |
Collapse
|
56
|
Schlingmann B, Molina SA, Koval M. Claudins: Gatekeepers of lung epithelial function. Semin Cell Dev Biol 2015; 42:47-57. [PMID: 25951797 DOI: 10.1016/j.semcdb.2015.04.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/24/2015] [Indexed: 12/25/2022]
Abstract
The lung must maintain a proper barrier between airspaces and fluid filled tissues in order to maintain lung fluid balance. Central to maintaining lung fluid balance are epithelial cells which create a barrier to water and solutes. The barrier function of these cells is mainly provided by tight junction proteins known as claudins. Epithelial barrier function varies depending on the different needs within the segments of the respiratory tree. In the lower airways, fluid is required to maintain mucociliary clearance, whereas in the terminal alveolar airspaces a thin layer of surfactant enriched fluid lowers surface tension to prevent airspace collapse and is critical for gas exchange. As the epithelial cells within the segments of the respiratory tree differ, the composition of claudins found in these epithelial cells is also different. Among these differences is claudin-18 which is uniquely expressed by the alveolar epithelial cells. Other claudins, notably claudin-4 and claudin-7, are more ubiquitously expressed throughout the respiratory epithelium. Claudin-5 is expressed by both pulmonary epithelial and endothelial cells. Based on in vitro and in vivo model systems and histologic analysis of lungs from human patients, roles for specific claudins in maintaining barrier function and protecting the lung from the effects of acute injury and disease are being identified. One surprising finding is that claudin-18 and claudin-4 control lung cell phenotype and inflammation beyond simply maintaining a selective paracellular permeability barrier. This suggests claudins have more nuanced roles for the control of airway and alveolar physiology in the healthy and diseased lung.
Collapse
Affiliation(s)
- Barbara Schlingmann
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Samuel A Molina
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Michael Koval
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, United States.
| |
Collapse
|
57
|
Akasaka K, Tanaka T, Maruyama T, Kitamura N, Hashimoto A, Ito Y, Watanabe H, Wakayama T, Arai T, Hayashi M, Moriyama H, Uchida K, Ohkouchi S, Tazawa R, Takada T, Yamaguchi E, Ichiwata T, Hirose M, Arai T, Inoue Y, Kobayashi H, Nakata K. A mathematical model to predict protein wash out kinetics during whole-lung lavage in autoimmune pulmonary alveolar proteinosis. Am J Physiol Lung Cell Mol Physiol 2015; 308:L105-17. [PMID: 25398988 DOI: 10.1152/ajplung.00239.2014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Whole-lung lavage (WLL) remains the standard therapy for pulmonary alveolar proteinosis (PAP), a process in which accumulated surfactants are washed out of the lung with 0.5-2.0 l of saline aliquots for 10-30 wash cycles. The method has been established empirically. In contrast, the kinetics of protein transfer into the lavage fluid has not been fully evaluated either theoretically or practically. Seventeen lungs from patients with autoimmune PAP underwent WLL. We made accurate timetables for each stage of WLL, namely, instilling, retaining, draining, and preparing. Subsequently, we measured the volumes of both instilled saline and drained lavage fluid, as well as the concentrations of proteins in the drained lavage fluid. We also proposed a mathematical model of protein transfer into the lavage fluid in which time is a single variable as the protein moves in response to the simple diffusion. The measured concentrations of IgG, transferrin, albumin, and β2-microglobulin closely matched the corresponding theoretical values calculated through differential equations. Coefficients for transfer of β2-microglobulin from the blood to the lavage fluid were two orders of magnitude higher than those of IgG, transferrin, and albumin. Simulations using the mathematical model showed that the cumulative amount of eliminated protein was not affected by the duration of each cycle but dependent mostly on the total time of lavage and partially on the volume instilled. Although physicians have paid little attention to the transfer of substances from the lung to lavage fluid, WLL seems to be a procedure that follows a diffusion-based mathematical model.
Collapse
Affiliation(s)
- Keiichi Akasaka
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Takahiro Tanaka
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Takashi Maruyama
- Disaster Prevention Research Institute, Kyoto University, Kyoto, Japan
| | - Nobutaka Kitamura
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Atsushi Hashimoto
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Yuko Ito
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Hiroyoshi Watanabe
- Department of Respiratory Medicine, Dokkyo Medical University Koshigaya Hospital, Saitama, Japan
| | - Tomoshige Wakayama
- Department of Respiratory Medicine, Dokkyo Medical University Koshigaya Hospital, Saitama, Japan
| | - Takero Arai
- Department of Anesthesiology, Dokkyo Medical University Koshigaya Hospital, Saitama, Japan
| | - Masachika Hayashi
- Division of Respiratory Medicine, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Hiroshi Moriyama
- Division of Respiratory Medicine, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Kanji Uchida
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinya Ohkouchi
- Department of Respiratory Medicine, Tohoku University Graduate school of Medicine, Miyagi, Japan
| | - Ryushi Tazawa
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Toshinori Takada
- Uonuma Institute of Community Medicine, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Etsuro Yamaguchi
- Department of Respiratory and Allergy Medicine, Aichi Medical University, Aichi, Japan
| | - Toshio Ichiwata
- Department of Pulmonary Medicine, Tokyo Medical University Hachioji Medical Center, Tokyo, Japan
| | - Masaki Hirose
- Clinical Research Center, NHO Kinki-Chuo Chest Medical Center, Osaka, Japan; and
| | - Toru Arai
- Clinical Research Center, NHO Kinki-Chuo Chest Medical Center, Osaka, Japan; and
| | - Yoshikazu Inoue
- Clinical Research Center, NHO Kinki-Chuo Chest Medical Center, Osaka, Japan; and
| | - Hirosuke Kobayashi
- Graduate School of Medical Sciences, Kitasato University, Kanagawa, Japan
| | - Koh Nakata
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan;
| |
Collapse
|
58
|
|
59
|
Colvin RA, Tanwandee T, Piratvisuth T, Thongsawat S, Hui AJ, Zhang H, Ren H, Chen PJ, Chuang WL, Sobhonslidsuk A, Li R, Qi Y, Praestgaard J, Han Y, Xu J, Stein DS. Randomized, controlled pharmacokinetic and pharmacodynamic evaluation of albinterferon in patients with chronic hepatitis B infection. J Gastroenterol Hepatol 2015; 30:184-91. [PMID: 24995515 DOI: 10.1111/jgh.12671] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/16/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Albinterferon is a fusion of albumin and interferon-α2b developed to improve the pharmacokinetics, convenience, and potential efficacy of interferon-α for the treatment of chronic hepatitis infections. METHODS This open-label, randomized, active-controlled, multicenter study investigated the safety and efficacy of albinterferon in patients with chronic hepatitis B virus (HBV) infection who were e-antigen (HBeAg) positive. One hundred and forty-one patients received one of four albinterferon doses/regimens or pegylated-interferon-α2a. Primary efficacy outcomes were changes in serum HBeAg and antibody, HBV-DNA, and alanine aminotransferase. Principal safety outcomes were changes in laboratory values, pulmonary function, and adverse events. RESULTS The study was prematurely terminated as phase III trials in hepatitis C infection indicated noninferior efficacy but inferior safety compared with pegylated-interferon-α2a. Here, all treatment groups had a significant reduction in HBV-DNA from baseline. Reductions in HBV-DNA were not significantly different, except the 1200 μg every 4 weeks albinterferon dose which was inferior compared with pegylated-interferon-α2a. The serum alanine aminotransferase levels decreased in all arms. The per-patient incidence of adverse events was not significantly different for albinterferon (96.4-100%) and pegylated-interferon-α2a (93.1%). Total adverse events, however, were higher for albinterferon and correlated to dose. Decreased lung function was found in all arms (∼93% of patients), and was more common in some albinterferon groups. CONCLUSIONS Albinterferon doses with similar anti-HBV efficacy to pegylated-interferon-α2a had higher rates of certain adverse events, particularly changes in lung diffusion capacity (http://www.clinicaltrials.gov number NCT00964665).
Collapse
Affiliation(s)
- Richard A Colvin
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Takano M, Kawami M, Aoki A, Yumoto R. Receptor-mediated endocytosis of macromolecules and strategy to enhance their transport in alveolar epithelial cells. Expert Opin Drug Deliv 2014; 12:813-25. [DOI: 10.1517/17425247.2015.992778] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
61
|
Orme IM. Vaccines to prevent tuberculosis infection rather than disease: Physiological and immunological aspects. Tuberculosis (Edinb) 2014; 101:210-216. [PMID: 25500316 DOI: 10.1016/j.tube.2014.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/02/2014] [Accepted: 10/22/2014] [Indexed: 12/11/2022]
Abstract
There is increasing enthusiasm and optimism that a vaccine could be developed that prevents infection rather than disease. In this article I discuss the fact that despite this optimism nothing has been produced so far that seems to have this capability, and moreover even the borderline between when infection ends and disease begins has not even been defined. To be effective such a vaccine, or at least the immunity it would generate, would have to work within the confines of the pulmonary physiological systems, which are complex. To date much of the emphasis here has turned away from T cell mediated immunity and towards establishing specific antibodies in the lungs. Here, I argue that with the exception of a possible exclusionary function, most claims of a protective role of antibody are completely over-blown. Finally, even if we had a potential "anti-infection" vaccine, how would we test and validate it?
Collapse
Affiliation(s)
- Ian M Orme
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
62
|
Thorley AJ, Ruenraroengsak P, Potter TE, Tetley TD. Critical determinants of uptake and translocation of nanoparticles by the human pulmonary alveolar epithelium. ACS NANO 2014; 8:11778-89. [PMID: 25360809 PMCID: PMC4246006 DOI: 10.1021/nn505399e] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 10/31/2014] [Indexed: 05/18/2023]
Abstract
The ability to manipulate the size and surface properties of nanomaterials makes them a promising vector for improving drug delivery and efficacy. Inhalation is a desirable route of administration as nanomaterials preferentially deposit in the alveolar region, a large surface area for drug absorption. However, as yet, the mechanisms by which particles translocate across the alveolar epithelial layer are poorly understood. Here we show that human alveolar type I epithelial cells internalize nanoparticles, whereas alveolar type II epithelial cells do not, and that nanoparticles translocate across the epithelial monolayer but are unable to penetrate the tight junctions between cells, ruling out paracellular translocation. Furthermore, using siRNA, we demonstrate that 50 nm nanoparticles enter largely by passive diffusion and are found in the cytoplasm, whereas 100 nm nanoparticles enter primarily via clathrin- and also caveolin-mediated endocytosis and are found in endosomes. Functionalization of nanoparticles increases their uptake and enhances binding of surfactant which further promotes uptake. Thus, we demonstrate that uptake and translocation across the pulmonary epithelium is controlled by alveolar type I epithelial cells, and furthermore, we highlight a number of factors that should be considered when designing new nanomedicines in order to improve drug delivery to the lung.
Collapse
Affiliation(s)
- Andrew J. Thorley
- Lung Cell Biology, Section of Pharmacology and Toxicology, National Heart and Lung Institute, Imperial College London, London SW3 6LY, U.K.
| | - Pakatip Ruenraroengsak
- Lung Cell Biology, Section of Pharmacology and Toxicology, National Heart and Lung Institute, Imperial College London, London SW3 6LY, U.K.
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
| | - Thomas E. Potter
- Lung Cell Biology, Section of Pharmacology and Toxicology, National Heart and Lung Institute, Imperial College London, London SW3 6LY, U.K.
| | - Teresa D. Tetley
- Lung Cell Biology, Section of Pharmacology and Toxicology, National Heart and Lung Institute, Imperial College London, London SW3 6LY, U.K.
| |
Collapse
|
63
|
Pfister T, Dolan D, Bercu J, Gould J, Wang B, Bechter R, Barle EL, Pfannkuch F, Flueckiger A. Bioavailability of therapeutic proteins by inhalation--worker safety aspects. ACTA ACUST UNITED AC 2014; 58:899-911. [PMID: 24958792 DOI: 10.1093/annhyg/meu038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
A literature review and analysis of inhalation bioavailability data for large therapeutic proteins was conducted in order to develop a practical estimate of the inhalation bioavailability of these drugs. This value is incorporated into equations used to derive occupational exposure limits(OELs) to protect biopharmaceutical manufacturing workers from systemic effects. Descriptive statistics implies that a value of 0.05, or 5% is an accurate estimate for large therapeutic proteins (molecular weight ≥ 40kDa). This estimate is confirmed by pharmacokinetic modeling of data from a human daily repeat-dose inhalation study of immunoglobulin G. In conclusion, we recommend using 5% bioavailability by inhalation when developing OELs for large therapeutic proteins.
Collapse
Affiliation(s)
- Thomas Pfister
- 1.F. Hoffmann - La Roche Ltd, Group Safety, Security, Health and Environmental Protection, CH-4070, Basel, Switzerland
| | - David Dolan
- 2.Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Joel Bercu
- 2.Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Janet Gould
- 3.Bristol-Myers Squibb Company New Brunswick, NJ 08903 , USA
| | - Bonnie Wang
- 3.Bristol-Myers Squibb Company New Brunswick, NJ 08903 , USA
| | | | | | - Friedlieb Pfannkuch
- 5.Roche Pharma Research and Early Development Department, Roche Innovation Center Basel, CH-4070 Basel, Switzerland
| | - Andreas Flueckiger
- 1.F. Hoffmann - La Roche Ltd, Group Safety, Security, Health and Environmental Protection, CH-4070, Basel, Switzerland
| |
Collapse
|
64
|
Iovino F, Molema G, Bijlsma JJE. Streptococcus pneumoniae Interacts with pIgR expressed by the brain microvascular endothelium but does not co-localize with PAF receptor. PLoS One 2014; 9:e97914. [PMID: 24841255 PMCID: PMC4026408 DOI: 10.1371/journal.pone.0097914] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 04/26/2014] [Indexed: 01/19/2023] Open
Abstract
Streptococcus pneumoniae is thought to adhere to the blood-brain barrier (BBB) endothelium prior to causing meningitis. The platelet activating factor receptor (PAFR) has been implicated in this adhesion but there is a paucity of data demonstrating direct binding of the bacteria to PAFR. Additionally, studies that inhibit PAFR strongly suggest that alternative receptors for pneumococci are present on the endothelium. Therefore, we studied the roles of PAFR and pIgR, an established epithelial pneumococcal receptor, in pneumococcal adhesion to brain endothelial cells in vivo. Mice were intravenously infected with pneumococci and sacrificed at various time points before meningitis onset. Co-localization of bacteria with PAFR and pIgR was investigated using immunofluorescent analysis of the brain tissue. In vitro blocking with antibodies and incubation of pneumococci with endothelial cell lysates were used to further probe bacteria-receptor interaction. In vivo as well as in vitro pneumococci did not co-localize with PAFR. On the other hand the majority of S. pneumoniae co-localized with endothelial pIgR and pIgR blocking reduced pneumococcal adhesion to endothelial cells. Pneumococci physically interacted with pIgR in endothelial cell lysates. In conclusion, bacteria did not associate with PAFR, indicating an indirect role of PAFR in pneumococcal adhesion to endothelial cells. In contrast, pIgR on the BBB endothelium may represent a novel pneumococcal adhesion receptor.
Collapse
Affiliation(s)
- Federico Iovino
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Grietje Molema
- Department of Pathology & Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jetta J. E. Bijlsma
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
65
|
Kaminskas LM, McLeod VM, Ryan GM, Kelly BD, Haynes JM, Williamson M, Thienthong N, Owen DJ, Porter CJH. Pulmonary administration of a doxorubicin-conjugated dendrimer enhances drug exposure to lung metastases and improves cancer therapy. J Control Release 2014; 183:18-26. [PMID: 24637466 DOI: 10.1016/j.jconrel.2014.03.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 02/24/2014] [Accepted: 03/09/2014] [Indexed: 11/27/2022]
Abstract
Direct administration of chemotherapeutic drugs to the lungs significantly enhances drug exposure to lung resident cancers and may improve chemotherapy when compared to intravenous administration. Direct inhalation of uncomplexed or unencapsulated cytotoxic drugs, however, leads to bolus release and unacceptable lung toxicity. Here, we explored the utility of a 56kDa PEGylated polylysine dendrimer, conjugated to doxorubicin, to promote the controlled and prolonged exposure of lung-resident cancers to cytotoxic drug. After intratracheal instillation to rats, approximately 60% of the dendrimer was rapidly removed from the lungs (within 24h) via mucociliary clearance and absorption into the blood. This was followed by a slower clearance phase that reflected both absorption from the lungs (bioavailability 10-13%) and biodegradation of the dendrimer scaffold. After 7days, approximately 15% of the dose remained in the lungs. A syngeneic rat model of lung metastasised breast cancer was subsequently employed to compare the anticancer activity of the dendrimer with a doxorubicin solution formulation after intravenous and pulmonary administration. Twice weekly intratracheal instillation of the dendrimer led to a >95% reduction in lung tumour burden after 2weeks in comparison to IV administration of doxorubicin solution which reduced lung tumour burden by only 30-50%. Intratracheal instillation of an equivalent dose of doxorubicin solution led to extensive lung-related toxicity and death withinseveral days of a single dose. The data suggest that PEGylated dendrimers have potential as inhalable drug delivery systems to promote the prolonged exposure of lung-resident cancers to chemotherapeutic drugs and to improve anti-cancer activity.
Collapse
Affiliation(s)
- Lisa M Kaminskas
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville, VIC 3052, Australia.
| | - Victoria M McLeod
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville, VIC 3052, Australia
| | - Gemma M Ryan
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville, VIC 3052, Australia
| | - Brian D Kelly
- Starpharma Pty Ltd, 75 Commercial Rd, Melbourne VIC 3004, Australia
| | - John M Haynes
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville VIC 3052, Australia
| | - Mark Williamson
- Gribbles Veterinary Pathology, 1868 Dandenong Rd, Clayton VIC 3168, Australia
| | | | - David J Owen
- Starpharma Pty Ltd, 75 Commercial Rd, Melbourne VIC 3004, Australia
| | - Christopher J H Porter
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville, VIC 3052, Australia.
| |
Collapse
|
66
|
Novak P, Shevchuk A, Ruenraroengsak P, Miragoli M, Thorley AJ, Klenerman D, Lab MJ, Tetley TD, Gorelik J, Korchev YE. Imaging single nanoparticle interactions with human lung cells using fast ion conductance microscopy. NANO LETTERS 2014; 14:1202-1207. [PMID: 24555574 DOI: 10.1021/nl404068p] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Experimental data on dynamic interactions between individual nanoparticles and membrane processes at nanoscale, essential for biomedical applications of nanoparticles, remain scarce due to limitations of imaging techniques. We were able to follow single 200 nm carboxyl-modified particles interacting with identified membrane structures at the rate of 15 s/frame using a scanning ion conductance microscope modified for simultaneous high-speed topographical and fluorescence imaging. The imaging approach demonstrated here opens a new window into the complexity of nanoparticle-cell interactions.
Collapse
Affiliation(s)
- Pavel Novak
- School of Engineering and Materials Science, Queen Mary University of London , Mile End Rd, London E1 4NS, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Goetzman ES, Alcorn JF, Bharathi SS, Uppala R, McHugh KJ, Kosmider B, Chen R, Zuo YY, Beck ME, McKinney RW, Skilling H, Suhrie KR, Karunanidhi A, Yeasted R, Otsubo C, Ellis B, Tyurina YY, Kagan VE, Mallampalli RK, Vockley J. Long-chain acyl-CoA dehydrogenase deficiency as a cause of pulmonary surfactant dysfunction. J Biol Chem 2014; 289:10668-10679. [PMID: 24591516 DOI: 10.1074/jbc.m113.540260] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Long-chain acyl-CoA dehydrogenase (LCAD) is a mitochondrial fatty acid oxidation enzyme whose expression in humans is low or absent in organs known to utilize fatty acids for energy such as heart, muscle, and liver. This study demonstrates localization of LCAD to human alveolar type II pneumocytes, which synthesize and secrete pulmonary surfactant. The physiological role of LCAD and the fatty acid oxidation pathway in lung was subsequently studied using LCAD knock-out mice. Lung fatty acid oxidation was reduced in LCAD(-/-) mice. LCAD(-/-) mice demonstrated reduced pulmonary compliance, but histological examination of lung tissue revealed no obvious signs of inflammation or pathology. The changes in lung mechanics were found to be due to pulmonary surfactant dysfunction. Large aggregate surfactant isolated from LCAD(-/-) mouse lavage fluid had significantly reduced phospholipid content as well as alterations in the acyl chain composition of phosphatidylcholine and phosphatidylglycerol. LCAD(-/-) surfactant demonstrated functional abnormalities when subjected to dynamic compression-expansion cycling on a constrained drop surfactometer. Serum albumin, which has been shown to degrade and inactivate pulmonary surfactant, was significantly increased in LCAD(-/-) lavage fluid, suggesting increased epithelial permeability. Finally, we identified two cases of sudden unexplained infant death where no lung LCAD antigen was detectable. Both infants were homozygous for an amino acid changing polymorphism (K333Q). These findings for the first time identify the fatty acid oxidation pathway and LCAD in particular as factors contributing to the pathophysiology of pulmonary disease.
Collapse
Affiliation(s)
- Eric S Goetzman
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213.
| | - John F Alcorn
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Sivakama S Bharathi
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Radha Uppala
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Kevin J McHugh
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Beata Kosmider
- Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Rimei Chen
- Department of Mechanical Engineering, University of Hawaii at Manoa, Honolulu, Hawaii 96822
| | - Yi Y Zuo
- Department of Mechanical Engineering, University of Hawaii at Manoa, Honolulu, Hawaii 96822
| | - Megan E Beck
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Richard W McKinney
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Helen Skilling
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Kristen R Suhrie
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Anuradha Karunanidhi
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Renita Yeasted
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Chikara Otsubo
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Bryon Ellis
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260; Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260; Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Rama K Mallampalli
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; Medical Specialty Service Line, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania 15213
| | - Jerry Vockley
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
68
|
Wu B, Sun YN. Pharmacokinetics of Peptide-Fc fusion proteins. J Pharm Sci 2013; 103:53-64. [PMID: 24285510 DOI: 10.1002/jps.23783] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 01/11/2023]
Abstract
Peptide-Fc fusion proteins (or peptibodies) are chimeric proteins generated by fusing a biologically active peptide with the Fc-domain of immunoglobulin G. In this review, we describe recent studies that have evaluated the absorption, distribution, metabolism, and excretion characteristics of peptibodies. Key features of the pharmacokinetics of peptibodies include their extended half-life due to recycling by the neonatal Fc receptor (FcRn), a substantial contribution by renal excretion to total clearance and, for certain peptibodies, target-mediated drug disposition. The prolonged half-life of peptibodies permits less-frequent dose administration compared with small therapeutic peptides, thereby supporting patient convenience and compliance. Hence, a considerable number of peptibodies are currently in preclinical and clinical development. Investigation of the metabolism (biotransformation) of biologics is an evolving area of research: ligand-binding mass spectrometry techniques have been employed for the characterization of the peptibody romiplostim, providing a new approach to evaluation of the degradation products of biologics. Pharmacokinetic/pharmacodynamic modeling and simulation techniques have been used to predict the pharmacokinetics of peptibodies which can inform clinical decision-making, particularly selection of dosing regimens. This integrated review highlights the distinct pharmacokinetic characteristics of peptibodies and their influence on the drug development process for this emerging family of therapeutics.
Collapse
Affiliation(s)
- Benjamin Wu
- Department of Pharmacokinetics and Drug Metabolism, Quantitative Pharmacology Group, Amgen Inc, Thousand Oaks, California, 91320
| | | |
Collapse
|
69
|
Mammalian cells preferentially internalize hydrogel nanodiscs over nanorods and use shape-specific uptake mechanisms. Proc Natl Acad Sci U S A 2013; 110:17247-52. [PMID: 24101456 DOI: 10.1073/pnas.1305000110] [Citation(s) in RCA: 284] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Size, surface charge, and material compositions are known to influence cell uptake of nanoparticles. However, the effect of particle geometry, i.e., the interplay between nanoscale shape and size, is less understood. Here we show that when shape is decoupled from volume, charge, and material composition, under typical in vitro conditions, mammalian epithelial and immune cells preferentially internalize disc-shaped, negatively charged hydrophilic nanoparticles of high aspect ratios compared with nanorods and lower aspect-ratio nanodiscs. Endothelial cells also prefer nanodiscs, however those of intermediate aspect ratio. Interestingly, unlike nanospheres, larger-sized hydrogel nanodiscs and nanorods are internalized more efficiently than their smallest counterparts. Kinetics, efficiency, and mechanisms of uptake are all shape-dependent and cell type-specific. Although macropinocytosis is used by both epithelial and endothelial cells, epithelial cells uniquely internalize these nanoparticles using the caveolae-mediated pathway. Human umbilical vein endothelial cells, on the other hand, use clathrin-mediated uptake for all shapes and show significantly higher uptake efficiency compared with epithelial cells. Using results from both upright and inverted cultures, we propose that nanoparticle internalization is a complex manifestation of three shape- and size-dependent parameters: particle surface-to-cell membrane contact area, i.e., particle-cell adhesion, strain energy for membrane deformation, and sedimentation or local particle concentration at the cell membrane. These studies provide a fundamental understanding on how nanoparticle uptake in different mammalian cells is influenced by the nanoscale geometry and is critical for designing improved nanocarriers and predicting nanomaterial toxicity.
Collapse
|
70
|
Mechanisms of absorption and elimination of drugs administered by inhalation. Ther Deliv 2013; 4:1027-45. [PMID: 23919477 DOI: 10.4155/tde.13.67] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pulmonary drug delivery is an effective route for local or systemic drug administration. However, compared with other routes of administration, there is a scarcity of information on how drugs are absorbed from the lung. The different cell composition lining the airways and alveoli makes this task extremely complicated. Lung cell lines and primary culture cells are useful in studying the absorption mechanisms. However, it is imperative that these cell cultures express essential features required to study these mechanisms such as intact tight junctions and transporters. In vivo, the drug has to face defensive physical and immunological barriers such as mucociliary clearance and alveolar macrophages. Knowledge of the physicochemical properties of the drug and aerosol formulation is required. All of these factors interact together leading to either successful drug deposition followed by absorption or drug elimination. These aspects concerning drug transport in the lung are addressed in this review.
Collapse
|
71
|
Intracellular delivery of polymeric nanocarriers: a matter of size, shape, charge, elasticity and surface composition. Ther Deliv 2013; 4:705-23. [PMID: 23738668 DOI: 10.4155/tde.13.37] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent progress in drug discovery has enabled the targeting of specific intracellular molecules to achieve therapeutic effects. These next-generation therapeutics are often biologics that cannot enter cells by mere diffusion. Therefore, it is imperative that drug carriers are efficiently internalized by cells and reach specific target organelles before releasing their cargo. Nanoscale polymeric carriers are particularly suitable for such intracellular delivery. Although size and surface charge have been the most studied parameters for nanocarriers, it is now well appreciated that other properties, for example, particle shape, elasticity and surface composition, also play a critical role in their transport across physiological barriers. It is proposed that a multivariate design space that considers the interdependence of particle geometry with its mechanical and surface properties must be optimized to formulate drug nanocarriers for effective accumulation at target sites and efficient intracellular delivery.
Collapse
|
72
|
Herold S, Gabrielli NM, Vadász I. Novel concepts of acute lung injury and alveolar-capillary barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 2013; 305:L665-81. [PMID: 24039257 DOI: 10.1152/ajplung.00232.2013] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this review we summarize recent major advances in our understanding on the molecular mechanisms, mediators, and biomarkers of acute lung injury (ALI) and alveolar-capillary barrier dysfunction, highlighting the role of immune cells, inflammatory and noninflammatory signaling events, mechanical noxae, and the affected cellular and molecular entities and functions. Furthermore, we address novel aspects of resolution and repair of ALI, as well as putative candidates for treatment of ALI, including pharmacological and cellular therapeutic means.
Collapse
Affiliation(s)
- Susanne Herold
- Dept. of Internal Medicine, Justus Liebig Univ., Universities of Giessen and Marburg Lung Center, Klinikstrasse 33, 35392 Giessen, Germany.
| | | | | |
Collapse
|
73
|
Coué G, Hermanns I, Unger RE, Kirkpatrick CJ, Engbersen JFJ. Development and in vitro Evaluation of Antigen-Loaded Poly(amidoamine) Nanoparticles for Respiratory Epithelium Applications. ChemMedChem 2013; 8:1787-94. [DOI: 10.1002/cmdc.201300307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Indexed: 11/10/2022]
|
74
|
McElroy MC, Kirton C, Gliddon D, Wolff RK. Inhaled biopharmaceutical drug development: nonclinical considerations and case studies. Inhal Toxicol 2013; 25:219-32. [PMID: 23480198 DOI: 10.3109/08958378.2013.769037] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Biopharmaceuticals are complex molecules often manufactured from living systems and their specificity and novelty holds great promise for the treatment of chronic diseases for which there are currently no cures. The inhalation route of biopharmaceutical drug delivery is attractive because the large surface area of the lung, and close proximity of the alveolar and vascular systems, maximizes the potential for drug delivery to the lung and/or systemic circulation. In addition, costs of delivery to the patient are potentially much reduced, in comparison with parental administration, since inhalation is non-invasive and likely to promote patient compliance. However, in comparison with small molecule drug development, developing an inhaled biopharmaceutical that is effective and safe for human use is associated with many challenges. This review considers some general principles of drug delivery to lung and issues associated with the translation of proof of concept studies to toxicology safety studies (e.g. aerosol generation, species selection, exaggerated pharmacology, and immunogenicity). This review also presents a summary of nonclinical and clinical data from inhaled biopharmaceuticals which are either marketed for human use or in Phase II clinical trials (e.g. DNase, insulin, human growth hormone, vaccines, therapeutic plasmid DNA complexes).
Collapse
Affiliation(s)
- Mary C McElroy
- Charles River Laboratories, Preclinical Services, Edinburgh, UK.
| | | | | | | |
Collapse
|
75
|
Cellular Uptake Mechanism and Therapeutic Utility of a Novel Peptide in Targeted-Delivery of Proteins into Neuronal Cells. Pharm Res 2013; 30:2108-17. [DOI: 10.1007/s11095-013-1068-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/24/2013] [Indexed: 12/22/2022]
|
76
|
High resolution fluorescence microscopy evidence on the transport of immunoglobulins. Differences between mammalian IgG, F(abʹ)2 and avian IgY. Toxicon 2013; 63:7-18. [DOI: 10.1016/j.toxicon.2012.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 10/18/2012] [Accepted: 10/30/2012] [Indexed: 11/21/2022]
|
77
|
|
78
|
Saha K, Kim ST, Yan B, Miranda OR, Alfonso FS, Shlosman D, Rotello VM. Surface functionality of nanoparticles determines cellular uptake mechanisms in mammalian cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:300-305. [PMID: 22972519 PMCID: PMC4070423 DOI: 10.1002/smll.201201129] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 07/04/2012] [Indexed: 04/14/2023]
Abstract
Nanoparticles (NPs) are versatile scaffolds for numerous biomedical applications including drug delivery and bioimaging. The surface functionality of NPs essentially dictates intracellular NP uptake and controls their therapeutic action. Using several pharmacological inhibitors, it is demonstrated that the cellular uptake mechanisms of cationic gold NPs in both cancer (HeLa) and normal cells (MCF10A) strongly depend on the NP surface monolayer, and mostly involve caveolae and dynamin-dependent pathways as well as specific cell surface receptors (scavenger receptors). Moreover, these NPs show different uptake mechanisms in cancer and normal cells, providing an opportunity to develop NPs with improved selectivity for delivery applications.
Collapse
|
79
|
Yumoto R, Suzuka S, Nishimoto S, Nagai J, Takano M. Enhancing Effect of Poly(amino acid)s on Albumin Uptake in Human Lung Epithelial A549 Cells. Drug Metab Pharmacokinet 2013; 28:497-503. [DOI: 10.2133/dmpk.dmpk-13-rg-028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
80
|
Huang Y, Aydintug MK, Loomis J, Macleod MK, McKee AS, Kirchenbaum G, Jakubzick CV, Kedl RM, Sun D, Jacobelli J, O'Brien RL, Born WK. Antigen-specific regulation of IgE antibodies by non-antigen-specific γδ T cells. THE JOURNAL OF IMMUNOLOGY 2012; 190:913-21. [PMID: 23275606 DOI: 10.4049/jimmunol.1202230] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We re-examined the observation that γδ T cells, when transferred from mice tolerized to an inhaled conventional Ag, suppress the allergic IgE response to this Ag specifically. Using OVA and hen egg lysozyme in crisscross fashion, we confirmed the Ag-specific IgE-regulatory effect of the γδ T cells. Although only Vγ4(+) γδ T cells are regulators, the Ag specificity does not stem from specificity of their γδ TCRs. Instead, the Vγ4(+) γδ T cells failed to respond to either Ag, but rapidly acquired Ag-specific regulatory function in vivo following i.v. injection of non-T cells derived from the spleen of Ag-tolerized mice. This correlated with their in vivo Ag acquisition from i.v. injected Ag-loaded splenic non-T cells, and in vivo transfer of membrane label provided evidence for direct contact between the injected splenic non-T cells and the Vγ4(+) γδ T cells. Together, our data suggest that Ag itself, when acquired by γδ T cells, directs the specificity of their IgE suppression.
Collapse
Affiliation(s)
- Yafei Huang
- Integrated Department of Immunology, National Jewish Health, Denver, CO 80206, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Takano M, Horiuchi T, Nagai J, Yumoto R. Effect of cigarette smoke extract on insulin transport in alveolar epithelial cell line A549. Lung 2012; 190:651-9. [PMID: 22960792 DOI: 10.1007/s00408-012-9413-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 08/21/2012] [Indexed: 11/26/2022]
Abstract
BACKGROUND The main purpose of this study was to evaluate the effect of cigarette smoke extract (CSE) on insulin transport in alveolar epithelial cells. METHODS We first examined the effect of CSE pretreatment on cell viability, mRNA expression, and lamellar body structures in A549 cells. Then the effect of CSE pretreatment on FITC-insulin transport was examined. RESULTS When A549 cells were treated with 30 μg/ml of CSE for 48 h, the expression of some mRNAs abundantly expressed in type II alveolar epithelial cells such as surfactant protein B was significantly increased. Lamellar bodylike structures became more evident with CSE treatment. FITC-insulin uptake from the apical side and subsequent efflux to the basal side was enhanced by CSE treatment in A549 cells. The enhancing effect of CSE on FITC-insulin uptake was concentration-dependent and reversible. A concentration-dependent enhancing effect of CSE on FITC-insulin uptake was also observed in normal, primary cultured alveolar type II epithelial cells isolated from rats. CONCLUSIONS Treatment of A549 cells by CSE may direct the cells to a more type II-like phenotype. In accordance with this observation, FITC-insulin uptake was enhanced by CSE treatment. These results may partly explain the higher insulin absorption from the lung in smokers than in nonsmokers.
Collapse
Affiliation(s)
- Mikihisa Takano
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | | | | | | |
Collapse
|
82
|
Naota M, Shimada A, Morita T, Yamamoto Y, Inoue K, Takano H. Caveolae-mediated endocytosis of intratracheally instilled gold colloid nanoparticles at the air-blood barrier in mice. Toxicol Pathol 2012; 41:487-96. [PMID: 22918937 DOI: 10.1177/0192623312457271] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Endocytosis is the primary mechanism by which nanoparticles are translocated over the alveolar epithelium. The purpose of this study was to elucidate the association between endocytosis and the translocation of nanoparticles at the air-blood barrier (ABB). Gold colloid particles (diameter, 20 nm) were intratracheally instilled into male ICR mice. Fifteen minutes after instillation, localized accumulation of agglomerated gold particles was observed in the cytoplasm of macrophages, on the surface of alveolar epithelial cells (AECs), and in alveoli. Electron microscopy revealed particles in the vesicles of macrophages, on the surface of AECs, and in caveolae-like vesicles in type 1 AECs. Immunohistochemistry demonstrated positive immunolabeling for caveolin-1 in the ABB of untreated lungs as well as lungs treated with gold particles. Double immunofluorescence and immunoelectron microscopy revealed the presence of caveolin-1 in AECs in the untreated lungs. These results suggest that instilled gold colloid particles are internalized into the alveolar epithelium at the ABB by caveolae-mediated endocytosis, which is regarded as a physiological function of AECs.
Collapse
Affiliation(s)
- Misaki Naota
- Department of Veterinary Pathology, Tottori University, Tottori, Japan
| | | | | | | | | | | |
Collapse
|
83
|
Buchäckert Y, Rummel S, Vohwinkel CU, Gabrielli NM, Grzesik BA, Mayer K, Herold S, Morty RE, Seeger W, Vadász I. Megalin mediates transepithelial albumin clearance from the alveolar space of intact rabbit lungs. J Physiol 2012; 590:5167-81. [PMID: 22826129 DOI: 10.1113/jphysiol.2012.233403] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The alveolo-capillary barrier is effectively impermeable to large solutes such as proteins. A hallmark of acute lung injury/acute respiratory distress syndrome is the accumulation of protein-rich oedema fluid in the distal airspaces. Excess protein must be cleared from the alveolar space for recovery; however, the mechanisms of protein clearance remain incompletely understood. In intact rabbit lungs 29.8 ± 2.2% of the radio-labelled alveolar albumin was transported to the vascular compartment at 37°C within 120 min, as assessed by real-time measurement of 125I-albumin clearance from the alveolar space. At 4°C or 22°C significantly lower albumin clearance (3.7 ± 0.4 or 16.2 ± 1.1%, respectively) was observed. Deposition of a 1000-fold molar excess of unlabelled albumin into the alveolar space or inhibition of cytoskeletal rearrangement or clathrin-dependent endocytosis largely inhibited the transport of 125I-albumin to the vasculature, while administration of unlabelled albumin to the vascular space had no effect on albumin clearance. Furthermore, albumin uptake capacity was measured as about 0.37 mg ml−1 in cultured rat lung epithelial monolayers, further highlighting the (patho)physiological relevance of active alveolar epithelial protein transport. Moreover, gene silencing and pharmacological inhibition of the multi-ligand receptor megalin resulted in significantly decreased albumin binding and uptake in monolayers of primary alveolar type II and type I-like and cultured lung epithelial cells. Our data indicate that clearance of albumin from the distal air spaces is facilitated by an active, high-capacity, megalin-mediated transport process across the alveolar epithelium. Further understanding of this mechanism is of clinical importance, since an inability to clear excess protein from the alveolar space is associated with poor outcome in patients with acute lung injury/acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Yasmin Buchäckert
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center, Klinikstrasse 33, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Mitchell WA, Castells A, Lang PO, Matas E, Lapenna A, Aspinall R. Pulmonary delivery of interleukin-7 provides efficient and safe delivery to the aging immune system. Rejuvenation Res 2012; 15:414-22. [PMID: 22663183 DOI: 10.1089/rej.2011.1258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Age-associated atrophy of the thymus with coincident reduction in thymopoeisis, decline in thymic output, and subsequent immune dysfunction has been reversed by the use of interleukin-7 (IL-7). In the earlier studies and in clinical trials, delivery of IL-7 has been by multiple injections over several days to maintain effective activity levels in the tissues. This is unlikely to meet with high compliance rates in future clinical use, and so we tested alternate routes of delivery using a technique involving tagging IL-7 with fluorescent dye that emits in the near-infrared region and whose fluorescence can be visualized within the tissues of live animals. We have shown that intratracheal instillation, enabling transfer through the lungs, provides an effective route for delivering IL-7 into the bloodstream and from there into the tissues in older animals. Delivery is rapid and widespread tissue distribution is seen. Comparison of administration either subcutaneously or by instillation reveals that IL-7 delivery by the pulmonary route provides significantly greater transmission to lymphoid tissues when compared with injection. In functional assessment studies, pulmonary administration led to significantly improved intrathymic T cell development in older animals when compared with IL-7 delivered by injection. Furthermore, in these older animals, delivery of IL-7 by intratracheal instillation was not accompanied by any apparent adverse events when compared with controls receiving saline vehicle by instillation or animals receiving IL-7 by subcutaneous injection.
Collapse
Affiliation(s)
- Wayne A Mitchell
- Regenerative Medicine Group, Translational Medicine, Cranfield Health, Cranfield University, Cranfield, United Kingdom
| | | | | | | | | | | |
Collapse
|
85
|
Desai N. Challenges in development of nanoparticle-based therapeutics. AAPS JOURNAL 2012; 14:282-95. [PMID: 22407288 PMCID: PMC3326161 DOI: 10.1208/s12248-012-9339-4] [Citation(s) in RCA: 503] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 02/17/2012] [Indexed: 02/08/2023]
Abstract
In recent years, nanotechnology has been increasingly applied to the area of drug development. Nanoparticle-based therapeutics can confer the ability to overcome biological barriers, effectively deliver hydrophobic drugs and biologics, and preferentially target sites of disease. However, despite these potential advantages, only a relatively small number of nanoparticle-based medicines have been approved for clinical use, with numerous challenges and hurdles at different stages of development. The complexity of nanoparticles as multi-component three dimensional constructs requires careful design and engineering, detailed orthogonal analysis methods, and reproducible scale-up and manufacturing process to achieve a consistent product with the intended physicochemical characteristics, biological behaviors, and pharmacological profiles. The safety and efficacy of nanomedicines can be influenced by minor variations in multiple parameters and need to be carefully examined in preclinical and clinical studies, particularly in context of the biodistribution, targeting to intended sites, and potential immune toxicities. Overall, nanomedicines may present additional development and regulatory considerations compared with conventional medicines, and while there is generally a lack of regulatory standards in the examination of nanoparticle-based medicines as a unique category of therapeutic agents, efforts are being made in this direction. This review summarizes challenges likely to be encountered during the development and approval of nanoparticle-based therapeutics, and discusses potential strategies for drug developers and regulatory agencies to accelerate the growth of this important field.
Collapse
Affiliation(s)
- Neil Desai
- Strategic Platforms, Abraxis BioScience, 11755 Wilshire Blvd., Suite 2300, Los Angeles, California 90025, USA.
| |
Collapse
|
86
|
Yumoto R, Suzuka S, Oda K, Nagai J, Takano M. Endocytic uptake of FITC-albumin by human alveolar epithelial cell line A549. Drug Metab Pharmacokinet 2012; 27:336-43. [PMID: 22214936 DOI: 10.2133/dmpk.dmpk-11-rg-127] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The uptake mechanism of FITC-labeled albumin (FITC-albumin) was examined in human alveolar epithelial cell line A549. FITC-albumin uptake by A549 cells was time- and temperature-dependent, and was markedly suppressed at 4°C compared with that at 37°C. The uptake was saturable, and was mediated by a high-affinity, low-capacity system and by a low-affinity, high-capacity system. In the following experiments, we focused on the low-affinity system. FITC-albumin uptake was markedly inhibited by metabolic inhibitors and by a vacuolar H⁺-ATPase, bafilomycin A₁. The uptake was inhibited by clathrin-mediated endocytosis inhibitors (phenylarsine oxide and chlorpromazine). Potassium depletion and hypertonicity that inhibit clathrin-mediated endocytosis also decreased FITC-albumin uptake. On the other hand, caveolae-mediated endocytosis inhibitors (indomethacin and nystatin) did not affect FITC-albumin uptake. In addition, FITC-albumin uptake was inhibited by macropinocytosis inhibitors such as 5-(N-ethyl-N-isopropyl) amiloride. These results suggest that the low-affinity system of FITC-albumin uptake is mediated by endocytosis in A549 cells, predominantly via a clathrin-mediated pathway. Macropinocytosis, but not caveolae-mediated endocytosis, may also be involved. Considering our previous findings, albumin may be transported by a similar mechanism and/or pathway in rat and human alveolar epithelial cells.
Collapse
Affiliation(s)
- Ryoko Yumoto
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | |
Collapse
|
87
|
Moradi E, Vllasaliu D, Garnett M, Falcone F, Stolnik S. Ligand density and clustering effects on endocytosis of folate modified nanoparticles. RSC Adv 2012. [DOI: 10.1039/c2ra01168a] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
88
|
Oda K, Yumoto R, Nagai J, Katayama H, Takano M. Enhancement Effect of Poly(amino acid)s on Insulin Uptake in Alveolar Epithelial Cells. Drug Metab Pharmacokinet 2012; 27:570-8. [DOI: 10.2133/dmpk.dmpk-12-rg-002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
89
|
Roth M. Is there a regulatory role of immunoglobulins on tissue forming cells relevant in chronic inflammatory lung diseases? J Allergy (Cairo) 2011; 2011:721517. [PMID: 22121383 PMCID: PMC3216316 DOI: 10.1155/2011/721517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 08/29/2011] [Indexed: 11/17/2022] Open
Abstract
Epithelial cells, fibroblasts and smooth muscle cells together form and give structure to the airway wall. These three tissue forming cell types are structure giving elements and participate in the immune response to inhaled particles including allergens and dust. All three cell types actively contribute to the pathogenesis of chronic inflammatory lung diseases such as asthma and chronic obstructive pulmonary disease (COPD). Tissue forming cells respond directly to allergens through activated immunoglobulins which then bind to their corresponding cell surface receptors. It was only recently reported that allergens and particles traffic through epithelial cells without modification and bind to the immunoglobulin receptors on the surface of sub-epithelial mesenchymal cells. In consequence, these cells secrete pro-inflammatory cytokines, thereby extending the local inflammation. Furthermore, activation of the immunoglobulin receptors can induce proliferation and tissue remodeling of the tissue forming cells. New studies using anti-IgE antibody therapy indicate that the inhibition of immunoglobulins reduces the response of tissue forming cells. The unmeasured questions are: (i) why do tissue forming cells express immunoglobulin receptors and (ii) do tissue forming cells process immunoglobulin receptor bound particles? The focus of this review is to provide an overview of the expression and function of various immunoglobulin receptors.
Collapse
Affiliation(s)
- Michael Roth
- Pulmonary Cell Research, Department of Research and Pneumology, University Hospital Basel, 4031 Basel, Switzerland
| |
Collapse
|
90
|
Oda K, Yumoto R, Nagai J, Katayama H, Takano M. Mechanism underlying insulin uptake in alveolar epithelial cell line RLE-6TN. Eur J Pharmacol 2011; 672:62-9. [PMID: 22004610 DOI: 10.1016/j.ejphar.2011.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 09/28/2011] [Accepted: 10/02/2011] [Indexed: 10/16/2022]
Abstract
For the development of efficient pulmonary delivery systems for protein and peptide drugs, it is important to understand their transport mechanisms in alveolar epithelial cells. In this study, the uptake mechanism for FITC-insulin in cultured alveolar epithelial cell line RLE-6TN was elucidated. FITC-insulin uptake by RLE-6TN cells was time-dependent, temperature-sensitive, and concentration-dependent. The uptake was inhibited by metabolic inhibitors, cytochalasin D, clathrin-mediated endocytosis inhibitors, and dynasore, an inhibitor of dynamin GTPase. On the other hand, no inhibitory effect was observed with caveolae-mediated endocytosis inhibitors and a macropinocytosis inhibitor. Intracellular FITC-insulin was found to be partly transported to the basal side of the epithelial cell monolayers. In addition, colocalization of FITC-insulin and LysoTracker Red was observed on confocal laser scanning microscopy, indicating that FITC-insulin was partly targeted to lysosomes. In accordance with these findings, SDS-PAGE/fluoroimage analysis showed that intact FITC-insulin in the cells was eliminated with time. The possible receptor involved in FITC-insulin uptake by RLE-6TN cells was examined by using siRNA. Transfection of the cells with megalin or insulin receptor siRNA successfully reduced the corresponding mRNA expression. FITC-insulin uptake decreased on the transfection with insulin receptor siRNA, but not that with megalin siRNA. These results suggest that insulin is taken up through endocytosis in RLE-6TN cells, and after the endocytosis, the intracellular insulin is partly degraded in lysosomes and partly transported to the basal side. Insulin receptor, but not megalin, may be involved at least partly in insulin endocytosis in RLE-6TN cells.
Collapse
Affiliation(s)
- Keisuke Oda
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | | | | | | | | |
Collapse
|
91
|
|
92
|
Koval M, Billaud M, Straub AC, Johnstone SR, Zarbock A, Duling BR, Isakson BE. Spontaneous lung dysfunction and fibrosis in mice lacking connexin 40 and endothelial cell connexin 43. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2536-46. [PMID: 21641379 PMCID: PMC3124229 DOI: 10.1016/j.ajpath.2011.02.045] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2010] [Revised: 12/30/2010] [Accepted: 02/01/2011] [Indexed: 12/31/2022]
Abstract
Gap junction proteins (connexins) facilitate intercellular communication and serve several roles in regulation of tissue function and remodeling. To examine the physiologic effects of depleting two prominent endothelial connexins, Cx40 and Cx43, transgenic mice were generated by breeding Cx40-deficient mice (Cx40(-/-)) with a vascular endothelial cell (VEC)-specific Cx43-deficient mouse strain (VEC Cx43(-/-)) to produce double-connexin knockout mice (VEC Cx43(-/-)/Cx40(-/-)). The life span in VEC Cx43(-/-)/Cx40(-/-) mice was dramatically shortened, which correlated with severe spontaneous lung abnormalities as the mice aged including increased fibrosis, aberrant alveolar remodeling, and increased lung fibroblast content. Moreover, VEC Cx43(-/-)/Cx40(-/-) mice exhibited cardiac hypertrophy and hypertension. Because VEC Cx43(-/-)/Cx40(-/-) mice demonstrated phenotypic hallmarks that were remarkably similar to those in mice deficient in caveolin-1, pulmonary caveolin expression was examined. Lungs from VEC Cx43(-/-)/Cx40(-/-) mice demonstrated significantly decreased expression of caveolin-1 and caveolin-2. This suggests that expression of caveolin-1 may be linked to expression of Cx40 and endothelial Cx43. Moreover, the phenotype of caveolin-1(-/-) mice and VEC Cx43(-/-)/Cx40(-/-) mice may arise via a common mechanism.
Collapse
Affiliation(s)
- Michael Koval
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Marie Billaud
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Adam C. Straub
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Scott R. Johnstone
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Alexander Zarbock
- Department of Anesthesiology and Critical Care Medicine, University of Münster, Münster, Germany
- Max-Planck-Institute of Molecular Biomedicine, Münster, Germany
| | - Brian R. Duling
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
93
|
Mitchell LA, Overgaard CE, Ward C, Margulies SS, Koval M. Differential effects of claudin-3 and claudin-4 on alveolar epithelial barrier function. Am J Physiol Lung Cell Mol Physiol 2011; 301:L40-9. [PMID: 21515662 DOI: 10.1152/ajplung.00299.2010] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Alveolar barrier function depends critically on the claudin family tight junction proteins. Of the major claudins expressed by alveolar epithelial cells, claudin (Cldn)-3 and Cldn-4 are the most closely related by amino acid homology, yet they differ dramatically in the pattern of expression. Previously published reports have shown that Cldn-3 is predominantly expressed by type II alveolar epithelial cells; Cldn-4 is expressed throughout the alveolar epithelium and is specifically upregulated in response to acute lung injury. Using primary rat alveolar epithelial cells transduced with yellow fluorescent protein-tagged claudin constructs, we have identified roles for Cldn-3 and Cldn-4 in alveolar epithelial barrier function. Surprisingly, increasing expression of Cldn-3 decreased alveolar epithelial barrier function, as assessed by transepithelial resistance and dye flux measurements. Conversely, increasing Cldn-4 expression improved alveolar epithelial transepithelial resistance compared with control cells. Other alveolar epithelial tight junction proteins were largely unaffected by increased expression of Cldn-3 and Cldn-4. Taken together, these results demonstrate that, in the context of the alveolar epithelium, Cldn-3 and Cldn-4 have different effects on paracellular permeability, despite significant homology in their extracellular loop domains.
Collapse
Affiliation(s)
- Leslie A Mitchell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
94
|
Sukumaran S, Jusko WJ, Dubois DC, Almon RR. Light-dark oscillations in the lung transcriptome: implications for lung homeostasis, repair, metabolism, disease, and drug action. J Appl Physiol (1985) 2011; 110:1732-47. [PMID: 21436464 DOI: 10.1152/japplphysiol.00079.2011] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diurnal-nocturnal, or circadian-like, rhythms are 24-h variations in biological processes, evolved for the efficient functioning of living organisms. Such oscillations and their regulation in many peripheral tissues are still unclear. In this study, we used Affymetrix gene chips in a rich time-series experiment involving 54 animals killed at 18 time points within the 24-h cycle to examine light-dark cycle patterns of gene expression in rat lungs. Data mining identified 646 genes (represented by 1,006 probe sets) showing robust oscillations in expression in lung that were parsed into 8 distinct temporal clusters. Surprisingly, more than two-thirds of the probe sets showing cyclic expression peaked during the animal's light/inactive period. Six core clock genes and nine clock-related genes showed rhythmic oscillations in their expression in lung. Many of the genes that peaked during the inactive period included genes related to extracellular matrix, cytoskeleton, and protein processing and trafficking, which appear to be mainly involved in the repair and remodeling of the organ. Genes coding for growth factor ligands and their receptors, which play important roles in maintaining normal lung function, also showed rhythmic expression. In addition, genes involved in the metabolism and transport of endogenous compounds, xenobiotics, and therapeutic drugs, along with genes that are biomarkers or potential therapeutic targets for many lung diseases, also exhibited 24-h cyclic oscillations, suggesting an important role for such rhythms in regulating various aspects of the physiology and pathophysiology of lung.
Collapse
Affiliation(s)
- Siddharth Sukumaran
- Dept. of Biological Sciences, 107 Hochstetter Hall, State Univ. of New York at Buffalo, Buffalo, NY 14260, USA
| | | | | | | |
Collapse
|
95
|
Yacobi NR, Fazllolahi F, Kim YH, Sipos A, Borok Z, Kim KJ, Crandall ED. Nanomaterial interactions with and trafficking across the lung alveolar epithelial barrier: implications for health effects of air-pollution particles. AIR QUALITY, ATMOSPHERE, & HEALTH 2011; 4:65-78. [PMID: 25568662 PMCID: PMC4283834 DOI: 10.1007/s11869-010-0098-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Studies on the health effects of air-pollution particles suggest that injury may result from inhalation of airborne ultrafine particles (<100 nm in diameter). Engineered nanomaterials (<100 nm in at least one dimension) may also be harmful if inhaled. Nanomaterials deposited on the respiratory epithelial tract are thought to cross the air-blood barrier, especially via the expansive alveolar region, into the systemic circulation to reach end organs (e.g., myocardium, liver, pancreas, kidney, and spleen). Since ambient ultrafine particles are difficult to track, studies of defined engineered nanomaterials have been used to obtain valuable information on how nanomaterials interact with and traffic across the air-blood barrier of mammalian lungs. Since specific mechanistic information on how nanomaterials interact with the lung is difficult to obtain using in vivo or ex vivo lungs due to their complex anatomy, in vitro alveolar epithelial models have been of considerable value in determining nanomaterial-lung interactions. In this review, we provide information on mechanisms underlying lung alveolar epithelial injury caused by various nanomaterials and on nanomaterial trafficking across alveolar epithelium that may lead to end-organ injury.
Collapse
Affiliation(s)
- Nazanin R. Yacobi
- Will Rogers Institute Pulmonary Research Center, University of Southern California, Los Angeles, CA 90033, USA. Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90033, USA
| | - Farnoosh Fazllolahi
- Will Rogers Institute Pulmonary Research Center, University of Southern California, Los Angeles, CA 90033, USA. Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90033, USA
| | - Yong Ho Kim
- Will Rogers Institute Pulmonary Research Center, University of Southern California, Los Angeles, CA 90033, USA. Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Arnold Sipos
- Will Rogers Institute Pulmonary Research Center, University of Southern California, Los Angeles, CA 90033, USA. Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Zea Borok
- Will Rogers Institute Pulmonary Research Center, University of Southern California, Los Angeles, CA 90033, USA. Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA. Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Kwang-Jin Kim
- Will Rogers Institute Pulmonary Research Center, University of Southern California, Los Angeles, CA 90033, USA. Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA. Department of Physiology and Biophysics, University of Southern California, Los Angeles, CA 90033, USA. Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90033, USA. Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90033, USA
| | - Edward D. Crandall
- Will Rogers Institute Pulmonary Research Center, University of Southern California, Los Angeles, CA 90033, USA. Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA. Department of Medicine, University of Southern California, IRD 620, 2020 Zonal Avenue, Los Angeles, CA 90033, USA. Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA. Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
96
|
Andrade F, Videira M, Ferreira D, Sarmento B. Nanocarriers for pulmonary administration of peptides and therapeutic proteins. Nanomedicine (Lond) 2011; 6:123-41. [DOI: 10.2217/nnm.10.143] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Peptides and therapeutic proteins have been the target of intense research and development in recent years by the pharmaceutical and biotechnology industry. Preferably, they are administered through the parenteral route, which is associated with reduced patient compliance. Formulations for noninvasive administration of peptides and therapeutic proteins are currently being developed. Among them, inhalation appears as a promising alternative for the administration of such products. Several formulations for pulmonary delivery are in various stages of development. Despite positive results, conventional formulations have some limitations such as reduced bioavailability and side effects. Nanocarriers may be an alternative way to overcome the problems of conventional formulations. Some nanocarrier-based formulations of peptides and therapeutic proteins are currently under development. The results obtained are promising, revealing the usefulness of these systems in the delivery of such drugs.
Collapse
Affiliation(s)
- Fernanda Andrade
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua Aníbal Cunha 164 4050-047, Portugal
| | - Mafalda Videira
- iMed.UL – Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Portugal
| | - Domingos Ferreira
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua Aníbal Cunha 164 4050-047, Portugal
| | - Bruno Sarmento
- Centro de Investigação em Ciências da Saúde (CICS), Department of Pharmaceutical Sciences, Instituto Superior de Ciências da Saúde – Norte, Gandra, Portugal
| |
Collapse
|
97
|
Abstract
Targeted nanoparticles have the potential to improve drug delivery efficiencies by more than two orders of magnitude, from the ~ 0.1% which is common today. Most pharmacologically agents on the market today are small drug molecules, which diffuse across the body’s blood-tissue barriers and distribute not only into the lesion, but into almost all organs. Drug actions in the non-lesion organs are an inescapable part of the drug delivery principle, causing “side-effects” which limit the maximally tolerable doses and result in inadequate therapy of many lesions. Nanoparticles only cross barriers by design, so side-effects are not built into their mode of operation. Delivery rates of almost 90% have been reported. This review examines the significance of these statements and checks how far they need qualification. What type of targeting is required? Is a single targeting sufficient? What new types of clinical challenge, such as immunogenicity, might attend the use of targeted nanoparticles?
Collapse
|
98
|
Selg E, Acevedo F, Nybom R, Blomgren B, Ryrfeldt Å, Gerde P. Delivering Horseradish Peroxidase as a Respirable Powder to the Isolated, Perfused, and Ventilated Lung of the Rat: The Pulmonary Disposition of an Inhaled Model Biopharmaceutical. J Aerosol Med Pulm Drug Deliv 2010; 23:273-84. [DOI: 10.1089/jamp.2009.0790] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ewa Selg
- Inhalation Sciences Sweden AB, Stockholm, Sweden
| | | | - Rolf Nybom
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Bo Blomgren
- Safety Assessment, AstraZeneca R&D, Södertälje, Sweden
| | - Åke Ryrfeldt
- Division of Physiology, The National Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Per Gerde
- Inhalation Sciences Sweden AB, Stockholm, Sweden
- Division of Physiology, The National Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
99
|
Cheung CY, Li S, Chen D, Brace RA. Regulation of caveolin-1 expression and phosphorylation by VEGF in ovine amnion cells. Reprod Sci 2010; 17:1112-9. [PMID: 20720263 DOI: 10.1177/1933719110378175] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascular endothelial growth factor (VEGF) has been implicated in the regulation of vesicular transport of amniotic fluid via caveolae across the amnion. This study tested the hypothesis that VEGF regulates caveolar function by stimulating caveolin-1 expression and phosphorylation in ovine amniotic epithelial cells (oAECs). Using primary cultures of oAECs, caveolin-1 was identified by immunofluorescent staining. Caveolin-1 messenger RNA (mRNA) abundance was determined by Reverse Transcription-Polymerase Chain Reaction (RT-PCR) and protein by Western blotting. The effects of VEGF( 165) on caveolin-1 expression and phosphorylation were determined. Caveolin-1 immunoreactivity was detected in oAECs. In response to 10 ng/mL VEGF( 165), caveolin-1 mRNA levels increased whereas the protein levels were unaffected. Furthermore, VEGF stimulated caveolin-1 phosphorylation, an effect abrogated by the inhibition of c-Src protein kinase. These data suggest that VEGF upregulates caveolin-1 activity through c-Src signaling pathways. Our observations support the hypothesis that VEGF regulates amniotic fluid transport across the amnion by stimulating caveolin-1 activity to mediate caveolar function in amnion cells.
Collapse
Affiliation(s)
- Cecilia Y Cheung
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97201, USA.
| | | | | | | |
Collapse
|
100
|
Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Yuan Hsin H, Ingber DE. Reconstituting organ-level lung functions on a chip. Science 2010; 328:1662-8. [PMID: 20576885 PMCID: PMC8335790 DOI: 10.1126/science.1188302] [Citation(s) in RCA: 2510] [Impact Index Per Article: 179.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Here, we describe a biomimetic microsystem that reconstitutes the critical functional alveolar-capillary interface of the human lung. This bioinspired microdevice reproduces complex integrated organ-level responses to bacteria and inflammatory cytokines introduced into the alveolar space. In nanotoxicology studies, this lung mimic revealed that cyclic mechanical strain accentuates toxic and inflammatory responses of the lung to silica nanoparticles. Mechanical strain also enhances epithelial and endothelial uptake of nanoparticulates and stimulates their transport into the underlying microvascular channel. Similar effects of physiological breathing on nanoparticle absorption are observed in whole mouse lung. Mechanically active "organ-on-a-chip" microdevices that reconstitute tissue-tissue interfaces critical to organ function may therefore expand the capabilities of cell culture models and provide low-cost alternatives to animal and clinical studies for drug screening and toxicology applications.
Collapse
Affiliation(s)
- Dongeun Huh
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
- Vascular Biology Program, Departments of Pathology & Surgery, Children’s Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | - Benjamin D. Matthews
- Vascular Biology Program, Departments of Pathology & Surgery, Children’s Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | - Akiko Mammoto
- Vascular Biology Program, Departments of Pathology & Surgery, Children’s Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | - Martin Montoya-Zavala
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
- Vascular Biology Program, Departments of Pathology & Surgery, Children’s Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | - Hong Yuan Hsin
- Vascular Biology Program, Departments of Pathology & Surgery, Children’s Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
- Vascular Biology Program, Departments of Pathology & Surgery, Children’s Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|