1
|
Peng W, Giesbers KC, Šiborová M, Beugelink JW, Pronker MF, Schulte D, Hilkens J, Janssen BJ, Strijbis K, Snijder J. Reverse-engineering the anti-MUC1 antibody 139H2 by mass spectrometry-based de novo sequencing. Life Sci Alliance 2024; 7:e202302366. [PMID: 38508723 PMCID: PMC10955041 DOI: 10.26508/lsa.202302366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
Mucin 1 (MUC1) is a transmembrane mucin expressed at the apical surface of epithelial cells at mucosal surfaces. MUC1 has a barrier function against bacterial invasion and is well known for its aberrant expression and glycosylation in adenocarcinomas. The MUC1 extracellular domain contains a variable number of tandem repeats (VNTR) of 20 amino acids, which are heavily O-linked glycosylated. Monoclonal antibodies against the MUC1 VNTR are powerful research tools with applications in the diagnosis and treatment of MUC1-expressing cancers. Here, we report direct mass spectrometry-based sequencing of anti-MUC1 hybridoma-derived 139H2 IgG, enabling reverse-engineering of the functional recombinant monoclonal antibody. The crystal structure of the 139H2 Fab fragment in complex with the MUC1 epitope was solved, revealing the molecular basis of 139H2 binding specificity to MUC1 and its tolerance to O-glycosylation of the VNTR. The available sequence of 139H2 will allow further development of MUC1-related diagnostic, targeting, and treatment strategies.
Collapse
Affiliation(s)
- Weiwei Peng
- https://ror.org/04pp8hn57 Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Koen Cap Giesbers
- https://ror.org/04pp8hn57 Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Marta Šiborová
- https://ror.org/04pp8hn57 Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - J Wouter Beugelink
- https://ror.org/04pp8hn57 Structural Biochemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Matti F Pronker
- https://ror.org/04pp8hn57 Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Douwe Schulte
- https://ror.org/04pp8hn57 Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - John Hilkens
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Bert Jc Janssen
- https://ror.org/04pp8hn57 Structural Biochemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Karin Strijbis
- https://ror.org/04pp8hn57 Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Joost Snijder
- https://ror.org/04pp8hn57 Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
2
|
Zhou Y, Chen H, Zhong W, Tao YJ. Collagen and actin network mediate antiviral immunity against Orsay virus in C. elegans intestinal cells. PLoS Pathog 2024; 20:e1011366. [PMID: 38190406 PMCID: PMC10798621 DOI: 10.1371/journal.ppat.1011366] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 01/19/2024] [Accepted: 12/18/2023] [Indexed: 01/10/2024] Open
Abstract
C. elegans is a free-living nematode that is widely used as a small animal model for studying fundamental biological processes and disease mechanisms. Since the discovery of the Orsay virus in 2011, C. elegans also holds the promise of dissecting virus-host interaction networks and innate antiviral immunity pathways in an intact animal. Orsay virus primarily targets the worm intestine, causing enlarged intestinal lumen as well as visible changes to infected cells such as liquefaction of cytoplasm and convoluted apical border. Previous studies of Orsay virus identified that C. elegans is able to mount antiviral responses by DRH-1/RIG-I mediated RNA interference and Intracellular Pathogen Response, a uridylyltransferase that destabilizes viral RNAs by 3' end uridylation, and ubiquitin protein modifications and turnover. To comprehensively search for novel antiviral pathways in C. elegans, we performed genome-wide RNAi screens by bacterial feeding using existing bacterial RNAi libraries covering 94% of the entire genome. Out of the 106 potential antiviral gene hits identified, we investigated those in three new pathways: collagens, actin remodelers, and epigenetic regulators. By characterizing Orsay virus infection in RNAi and mutant worms, our results indicate that collagens likely form a physical barrier in intestine cells to inhibit viral infection by preventing Orsay virus entry. Furthermore, evidence suggests that actin remodeling proteins (unc-34, wve-1 and wsp-1) and chromatin remodelers (nurf-1 and isw-1) exert their antiviral activities by regulating the intestinal actin (act-5), a critical component of the terminal web which likely function as another physical barrier to prevent Orsay infection.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Hanqiao Chen
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Weiwei Zhong
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Yizhi Jane Tao
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| |
Collapse
|
3
|
Frey A, Lunding LP, Wegmann M. The Dual Role of the Airway Epithelium in Asthma: Active Barrier and Regulator of Inflammation. Cells 2023; 12:2208. [PMID: 37759430 PMCID: PMC10526792 DOI: 10.3390/cells12182208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic airway inflammation is the cornerstone on which bronchial asthma arises, and in turn, chronic inflammation arises from a complex interplay between environmental factors such as allergens and pathogens and immune cells as well as structural cells constituting the airway mucosa. Airway epithelial cells (AECs) are at the center of these processes. On the one hand, they represent the borderline separating the body from its environment in order to keep inner homeostasis. The airway epithelium forms a multi-tiered, self-cleaning barrier that involves an unstirred, discontinuous mucous layer, the dense and rigid mesh of the glycocalyx, and the cellular layer itself, consisting of multiple, densely interconnected cell types. On the other hand, the airway epithelium represents an immunologically highly active tissue once its barrier has been penetrated: AECs play a pivotal role in releasing protective immunoglobulin A. They express a broad spectrum of pattern recognition receptors, enabling them to react to environmental stressors that overcome the mucosal barrier. By releasing alarmins-proinflammatory and regulatory cytokines-AECs play an active role in the formation, strategic orientation, and control of the subsequent defense reaction. Consequently, the airway epithelium is of vital importance to chronic inflammatory diseases, such as asthma.
Collapse
Affiliation(s)
- Andreas Frey
- Division of Mucosal Immunology and Diagnostics, Research Center Borstel, 23845 Borstel, Germany;
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927 Großhansdorf, Germany;
| | - Lars P. Lunding
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927 Großhansdorf, Germany;
- Division of Lung Immunology, Research Center Borstel, 23845 Borstel, Germany
| | - Michael Wegmann
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927 Großhansdorf, Germany;
- Division of Lung Immunology, Research Center Borstel, 23845 Borstel, Germany
| |
Collapse
|
4
|
Yoo D, Whang CH, Hong J, Kim D, Prayogo MC, Son Y, Jung W, Lee S, Lee HS, Jon S. Anti-inflammatory Glycocalyx-Mimicking Nanoparticles for Colitis Treatment: Construction and In Vivo Evaluation. Angew Chem Int Ed Engl 2023; 62:e202304815. [PMID: 37310766 DOI: 10.1002/anie.202304815] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 06/14/2023]
Abstract
Common medications for treating inflammatory bowel disease (IBD) have limited therapeutic efficacy and severe adverse effects. This underscores the urgent need for novel therapeutic approaches that can effectively target inflamed sites in the gastrointestinal tract upon oral administration, exerting potent therapeutic efficacy while minimizing systemic effects. Here, we report the construction and in vivo therapeutic evaluation of a library of anti-inflammatory glycocalyx-mimicking nanoparticles (designated GlyNPs) in a mouse model of IBD. The anti-inflammatory GlyNP library was created by attaching bilirubin (BR) to a library of glycopolymers composed of random combinations of the five most naturally abundant sugars. Direct in vivo screening of 31 BR-attached anti-inflammatory GlyNPs via oral administration into mice with acute colitis led to identification of a candidate GlyNP capable of targeting macrophages in the inflamed colon and effectively alleviating colitis symptoms. These findings suggest that the BR-attached GlyNP library can be used as a platform to identify anti-inflammatory nanomedicines for various inflammatory diseases.
Collapse
Affiliation(s)
- Dohyun Yoo
- Department of Biological Sciences, Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Chang-Hee Whang
- Department of Biological Sciences, Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Jungwoo Hong
- Department of Chemistry, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Multiscale Chiral Architectures (CMCA), KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Dohyeon Kim
- Department of Biological Sciences, Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Monica Celine Prayogo
- Department of Biological Sciences, Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Youngju Son
- Department of Biological Sciences, Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Wonsik Jung
- Department of Biological Sciences, Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Seojung Lee
- Department of Biological Sciences, Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Hee-Seung Lee
- Department of Chemistry, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Multiscale Chiral Architectures (CMCA), KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences, Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| |
Collapse
|
5
|
Li S, Keenan JI, Shaw IC, Frizelle FA. Could Microplastics Be a Driver for Early Onset Colorectal Cancer? Cancers (Basel) 2023; 15:3323. [PMID: 37444433 DOI: 10.3390/cancers15133323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/12/2023] [Accepted: 06/18/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction: The incidence of colorectal cancer in those under 50 years of age (early onset colorectal cancer (EOCRC)) is increasing throughout the world. This has predominantly been an increase in distal colonic and rectal cancers, which are biologically similar to late onset colorectal cancer (LOCRC) but with higher rates of mucinous or signet ring histology, or poorly differentiated cancers. The epidemiology of this change suggests that it is a cohort effect since 1960, and is most likely driven by an environmental cause. We explore the possible role of microplastics as a driver for this change. Review: The development of sporadic colorectal cancer is likely facilitated by the interaction of gut bacteria and the intestinal wall. Normally, a complex layer of luminal mucus provides colonocytes with a level of protection from the effects of these bacteria and their toxins. Plastics were first developed in the early 1900s. After 1945 they became more widely used, with a resultant dramatic increase in plastic pollution and their breakdown to microplastics. Microplastics (MPs) are consumed by humans from an early age and in increasingly large quantities. As MPs pass through the gastrointestinal tract they interact with the normal physiological mechanism of the body, particularly in the colon and rectum, where they may interact with the protective colonic mucus layer. We describe several possible mechanisms of how microplastics may disrupt this mucus layer, thus reducing its protective effect and increasing the likelihood of colorectal cancer. Conclusions: The epidemiology of increase in EOCRC suggests an environmental driver. This increase in EOCRC matches the time sequence in which we could expect to see an effect of rapid increase of MPs in the environment and, as such, we have explored possible mechanisms for this effect. We suggest that it is possible that the MPs damage the barrier integrity of the colonic mucus layer, thus reducing its protective effect. MPs in CRC pathogenesis warrants further investigation. Future directions: Further clarification needs to be sought regarding the interaction between MPs, gut microbiota and the mucus layer. This will need to be modelled in long-term animal studies to better understand how chronic consumption of environmentally-acquired MPs may contribute to an increased risk of colorectal carcinogenesis.
Collapse
Affiliation(s)
- Shelley Li
- Department of Surgery, University of Otago Christchurch, Christchurch 8011, New Zealand
| | - Jacqueline I Keenan
- Department of Surgery, University of Otago Christchurch, Christchurch 8011, New Zealand
| | - Ian C Shaw
- School of Physical & Chemical Sciences, University of Canterbury, Christchurch 8041, New Zealand
| | - Frank A Frizelle
- Department of Surgery, University of Otago Christchurch, Christchurch 8011, New Zealand
| |
Collapse
|
6
|
Izadifar Z, Sontheimer-Phelps A, Lubamba BA, Bai H, Fadel C, Stejskalova A, Ozkan A, Dasgupta Q, Bein A, Junaid A, Gulati A, Mahajan G, Kim S, LoGrande NT, Naziripour A, Ingber DE. Modeling mucus physiology and pathophysiology in human organs-on-chips. Adv Drug Deliv Rev 2022; 191:114542. [PMID: 36179916 DOI: 10.1016/j.addr.2022.114542] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 08/25/2022] [Accepted: 09/13/2022] [Indexed: 01/24/2023]
Abstract
The surfaces of human internal organs are lined by a mucus layer that ensures symbiotic relationships with commensal microbiome while protecting against potentially injurious environmental chemicals, toxins, and pathogens, and disruption of this layer can contribute to disease development. Studying mucus biology has been challenging due to the lack of physiologically relevant human in vitro models. Here we review recent progress that has been made in the development of human organ-on-a-chip microfluidic culture models that reconstitute epithelial tissue barriers and physiologically relevant mucus layers with a focus on lung, colon, small intestine, cervix and vagina. These organ-on-a-chip models that incorporate dynamic fluid flow, air-liquid interfaces, and physiologically relevant mechanical cues can be used to study mucus composition, mechanics, and structure, as well as investigate its contributions to human health and disease with a level of biomimicry not possible in the past.
Collapse
Affiliation(s)
- Zohreh Izadifar
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | | | - Bob A Lubamba
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Haiqing Bai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Cicely Fadel
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Anna Stejskalova
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Alican Ozkan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Queeny Dasgupta
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Amir Bein
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Abidemi Junaid
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Aakanksha Gulati
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Gautam Mahajan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Seongmin Kim
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Nina T LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Arash Naziripour
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States; Vascular Biology Program, Boston Children's Hospital and Department of Pathology, Harvard Medical School, Boston, MA 02115, United States; Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA 02138, United Kingdom.
| |
Collapse
|
7
|
Kim H, Rahmawati L, Hong YH, Choi SY, Cho JY. NK cell-mediated immunostimulatory effects of ethanol extract of Morinda citrifolia (noni) fruit. BMC Complement Med Ther 2022; 22:222. [PMID: 35996139 PMCID: PMC9394078 DOI: 10.1186/s12906-022-03700-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/27/2022] [Indexed: 11/10/2022] Open
Abstract
Background Morinda citrifolia (Noni) is a plant that has long been used in various products such as foods and cosmetics. Although noni has been known to have immunostimulatory activity, detailed mechanism at the cellular level has not been fully elucidated yet. In this study, we focused on understanding as to how noni fruit can positively stimulate body’s immune responses. Methods To do this, an ethanol extract of noni fruit (Mc-fEE) was prepared and administered for 30 days to male C57BL/6 mice for in vivo experiment. NK cell activity and cytokine production level from Mc-fEE-treated mice were analyzed by flowcytometry, real-time PCR, and ELISA. Mc-fEE-triggered molecular events were detected from RAW264.7 cells and splenocytes using Western blotting and real-time PCR analyses. Results The mRNA expression levels of cytokines such as interleukin families, interferon (IFN)-β, and tumor necrosis factor (TNF)-α were increased by Mc-fEE treatment in vitro and in vivo. Western blotting analysis showed that the phosphorylation levels of nuclear factor (NF)-κB and activator protein (AP)-1 subunits these were enhanced in Mc-fEE-treated RAW264.7 cells. In addition, according to in vivo experiments, it was considered that Mc-fEE can increase the population of splenic NK cells and subsequent upregulation of their cytotoxic activity against YAC-1 cells, a T- cell lymphoma. Conclusion In this paper, we could confirm that Mc-fEE has remarkable immunostimulatory effects by activation and increase of the NK cell population. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03700-3.
Collapse
|
8
|
Ménard S, Lacroix-Lamandé S, Ehrhardt K, Yan J, Grassl GA, Wiedemann A. Cross-Talk Between the Intestinal Epithelium and Salmonella Typhimurium. Front Microbiol 2022; 13:906238. [PMID: 35733975 PMCID: PMC9207452 DOI: 10.3389/fmicb.2022.906238] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Salmonella enterica serovars are invasive gram-negative bacteria, causing a wide range of diseases from gastroenteritis to typhoid fever, representing a public health threat around the world. Salmonella gains access to the intestinal lumen after oral ingestion of contaminated food or water. The crucial initial step to establish infection is the interaction with the intestinal epithelium. Human-adapted serovars such as S. Typhi or S. Paratyphi disseminate to systemic organs and induce life-threatening disease known as typhoid fever, whereas broad-host serovars such as S. Typhimurium usually are limited to the intestine and responsible for gastroenteritis in humans. To overcome intestinal epithelial barrier, Salmonella developed mechanisms to induce cellular invasion, intracellular replication and to face host defence mechanisms. Depending on the serovar and the respective host organism, disease symptoms differ and are linked to the ability of the bacteria to manipulate the epithelial barrier for its own profit and cross the intestinal epithelium.This review will focus on S. Typhimurium (STm). To better understand STm pathogenesis, it is crucial to characterize the crosstalk between STm and the intestinal epithelium and decipher the mechanisms and epithelial cell types involved. Thus, the purpose of this review is to summarize our current knowledge on the molecular dialogue between STm and the various cell types constituting the intestinal epithelium with a focus on the mechanisms developed by STm to cross the intestinal epithelium and access to subepithelial or systemic sites and survive host defense mechanisms.
Collapse
Affiliation(s)
- Sandrine Ménard
- IRSD - Institut de Recherche en Santé Digestive, Université́ de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | | | - Katrin Ehrhardt
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Jin Yan
- IRSD - Institut de Recherche en Santé Digestive, Université́ de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| | - Guntram A. Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Agnès Wiedemann
- IRSD - Institut de Recherche en Santé Digestive, Université́ de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- *Correspondence: Agnès Wiedemann,
| |
Collapse
|
9
|
Crowe TP, Hsu WH. Evaluation of Recent Intranasal Drug Delivery Systems to the Central Nervous System. Pharmaceutics 2022; 14:629. [PMID: 35336004 PMCID: PMC8950509 DOI: 10.3390/pharmaceutics14030629] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
Neurological diseases continue to increase in prevalence worldwide. Combined with the lack of modifiable risk factors or strongly efficacious therapies, these disorders pose a significant and growing burden on healthcare systems and societies. The development of neuroprotective or curative therapies is limited by a variety of factors, but none more than the highly selective blood-brain barrier. Intranasal administration can bypass this barrier completely and allow direct access to brain tissues, enabling a large number of potential new therapies ranging from bioactive peptides to stem cells. Current research indicates that merely administering simple solutions is inefficient and may limit therapeutic success. While many therapies can be delivered to some degree without carrier molecules or significant modification, a growing body of research has indicated several methods of improving the safety and efficacy of this administration route, such as nasal permeability enhancers, gelling agents, or nanocarrier formulations. This review shall discuss promising delivery systems and their role in expanding the clinical efficacy of this novel administration route. Optimization of intranasal administration will be crucial as novel therapies continue to be studied in clinical trials and approved to meet the growing demand for the treatment of patients with neurological diseases.
Collapse
Affiliation(s)
- Tyler P. Crowe
- Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Walter H. Hsu
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
10
|
Chaikhumwang P, Madapong A, Saeng-Chuto K, Nilubol D, Tantituvanont A. Intranasal delivery of inactivated PRRSV loaded cationic nanoparticles coupled with enterotoxin subunit B induces PRRSV-specific immune responses in pigs. Sci Rep 2022; 12:3725. [PMID: 35260663 PMCID: PMC8904483 DOI: 10.1038/s41598-022-07680-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/08/2022] [Indexed: 12/18/2022] Open
Abstract
This study was conducted to evaluate the induction of systemic and mucosal immune responses and protective efficacy following the intranasal administration of inactivated porcine reproductive and respiratory syndrome virus (PRRSV) loaded in polylactic acid (PLA) nanoparticles coupled with heat-labile enterotoxin subunit B (LTB) and dimethyldioctadecylammonium bromide (DDA). Here, 42- to 3-week-old PRRSV-free pigs were randomly allocated into 7 groups of 6 pigs each. Two groups represented the negative (nonvaccinated pigs/nonchallenged pigs, NoVacNoChal) and challenge (nonvaccinated/challenged, NoVacChal) controls. The pigs in the other 5 groups, namely, PLA nanoparticles/challenged (blank NPs), LTB-DDA coupled with PLA nanoparticles/challenged (adjuvant-blank NPs), PLA nanoparticles-encapsulating inactivated PRRSV/challenged (KNPs), LTB-DDA coupled with PLA nanoparticles loaded with inactivated PRRSV/challenged pigs (adjuvant-KNPs) and inactivated PRRSV/challenged pigs (inactivated PRRSV), were intranasally vaccinated with previously described vaccines at 0, 7 and 14 days post-vaccination (DPV). Serum and nasal swab samples were collected weekly and assayed by ELISA to detect the presence of IgG and IgA, respectively. Viral neutralizing titer (VNT) in sera, IFN-γ-producing cells and IL-10 secretion in stimulated peripheral blood mononuclear cells (PBMCs) were also measured. The pigs were intranasally challenged with PRRSV-2 at 28 DPV and necropsied at 35 DPV, and then macro- and microscopic lung lesions were evaluated. The results demonstrated that following vaccination, adjuvant-KNP-vaccinated pigs had significantly higher levels of IFN-γ-producing cells, VNT and IgG in sera, and IgA in nasal swab samples and significantly lower IL-10 levels than the other vaccinated groups. Following challenge, the adjuvant-KNP-vaccinated pigs had significantly lower PRRSV RNA and macro- and microscopic lung lesions than the other vaccinated groups. In conclusion, the results of the study demonstrated that adjuvant-KNPs are effective in eliciting immune responses against PRRSV and protecting against PRRSV infections over KNPs and inactivated PRRSV and can be used as an adjuvant for intranasal PRRSV vaccines.
Collapse
Affiliation(s)
- Puwich Chaikhumwang
- Division of Pharmaceutical Sciences, Department of Pharmaceutical Care, Faculty of Pharmaceutical Sciences, University of Phayao, Phayao, 56000, Thailand
| | - Adthakorn Madapong
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kepalee Saeng-Chuto
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Dachrit Nilubol
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Angkana Tantituvanont
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
11
|
Nanomaterial-Induced Extra-Pulmonary Health Effects – the Importance of Next Generation Physiologically Relevant In Vitro Test Systems for the Future of Nanotoxicology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1357:259-273. [DOI: 10.1007/978-3-030-88071-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
Miao YB, Lin YJ, Chen KH, Luo PK, Chuang SH, Yu YT, Tai HM, Chen CT, Lin KJ, Sung HW. Engineering Nano- and Microparticles as Oral Delivery Vehicles to Promote Intestinal Lymphatic Drug Transport. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2104139. [PMID: 34596293 DOI: 10.1002/adma.202104139] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/20/2021] [Indexed: 06/13/2023]
Abstract
Targeted oral delivery of a drug via the intestinal lymphatic system (ILS) has the advantages of protecting against hepatic first-pass metabolism of the drug and improving its pharmacokinetic performance. It is also a promising route for the oral delivery of vaccines and therapeutic agents to induce mucosal immune responses and treat lymphatic diseases, respectively. This article describes the anatomical structures and physiological characteristics of the ILS, with an emphasis on enterocytes and microfold (M) cells, which are the main gateways for the transport of particulate delivery vehicles across the intestinal epithelium into the lymphatics. A comprehensive overview of recent advances in the rational engineering of particulate vehicles, along with the challenges and opportunities that they present for improving ILS drug delivery, is provided, and the mechanisms by which such vehicles target and transport through enterocytes or M cells are discussed. The use of naturally sourced materials, such as yeast microcapsules and their derived polymeric β-glucans, as novel ILS-targeting delivery vehicles is also reviewed. Such use is the focus of an emerging field of research. Their potential use in the oral delivery of nucleic acids, such as mRNA vaccines, is proposed.
Collapse
Affiliation(s)
- Yang-Bao Miao
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Yu-Jung Lin
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Kuan-Hung Chen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Po-Kai Luo
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Shun-Hao Chuang
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Yu-Tzu Yu
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Hsien-Meng Tai
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan, Republic of China
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Molecular Imaging Center, Linkou Chang Gung Memorial Hospital, and Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| |
Collapse
|
13
|
Spliid CB, Toledo AG, Sanderson P, Mao Y, Gatto F, Gustavsson T, Choudhary S, Saldanha AL, Vogelsang RP, Gögenur I, Theander TG, Leach FE, Amster IJ, Esko JD, Salanti A, Clausen TM. The specificity of the malarial VAR2CSA protein for chondroitin sulfate depends on 4-O-sulfation and ligand accessibility. J Biol Chem 2021; 297:101391. [PMID: 34762909 DOI: 10.1016/j.jbc.2021.101391] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022] Open
Abstract
Placental malaria infection is mediated by the binding of the malarial VAR2CSA protein to the placental glycosaminoglycan, chondroitin sulfate. Recombinant sub-fragments of VAR2CSA (rVAR2) have also been shown to bind specifically and with high affinity to cancer cells and tissues, suggesting the presence of a shared type of oncofetal chondroitin sulfate (ofCS) in the placenta and in tumors. However, the exact structure of ofCS and what determines the selective tropism of VAR2CSA remains poorly understood. In this study, ofCS was purified by affinity chromatography using rVAR2 and subjected to detailed structural analysis. We found high levels of N-acetylgalactosamine 4-O-sulfation (∼80-85%) in placenta- and tumor-derived ofCS. This level of 4-O-sulfation was also found in other tissues that do not support parasite sequestration, suggesting that VAR2CSA tropism is not exclusively determined by placenta- and tumor-specific sulfation. Here, we show that both placenta and tumors contain significantly more chondroitin sulfate moieties of higher molecular weight than other tissues. In line with this, CHPF and CHPF2, which encode proteins required for chondroitin polymerization, are significantly upregulated in most cancer types. CRISPR/Cas9 targeting of CHPF and CHPF2 in tumor cells reduced the average molecular weight of cell-surface chondroitin sulfate and resulted in a marked reduction of rVAR2 binding. Finally, utilizing a cell-based glycocalyx model, we showed that rVAR2 binding correlates with the length of the chondroitin sulfate chains in the cellular glycocalyx. These data demonstrate that the total amount and cellular accessibility of chondroitin sulfate chains impact rVAR2 binding and thus malaria infection.
Collapse
Affiliation(s)
- Charlotte B Spliid
- Department of Cellular and Molecular Medicine, and Glycobiology Research and Training Center, University of California San Diego, La Jolla, CA, USA; Centre for Medical Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Alejandro Gomez Toledo
- Department of Cellular and Molecular Medicine, and Glycobiology Research and Training Center, University of California San Diego, La Jolla, CA, USA; Department of Clinical Sciences, Division of Infection Medicine, Lund University, Sweden
| | | | - Yang Mao
- School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, China and Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, 510990 Guangzhou, China
| | - Francesco Gatto
- Department of Biology and Biological Engineering, Chalmers University of Technology, 42196 Gothenburg, Sweden
| | - Tobias Gustavsson
- Centre for Medical Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Swati Choudhary
- Centre for Medical Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Ana L Saldanha
- Centre for Medical Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Rasmus P Vogelsang
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, DK-4600 Koege, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, DK-4600 Koege, Denmark
| | - Thor G Theander
- Centre for Medical Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Franklin E Leach
- Department of Environmental Health Science, University of Georgia, Athens, GA 30602
| | | | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, and Glycobiology Research and Training Center, University of California San Diego, La Jolla, CA, USA
| | - Ali Salanti
- Centre for Medical Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Thomas Mandel Clausen
- Department of Cellular and Molecular Medicine, and Glycobiology Research and Training Center, University of California San Diego, La Jolla, CA, USA; Centre for Medical Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200 Copenhagen, Denmark.
| |
Collapse
|
14
|
Fat of the Gut: Epithelial Phospholipids in Inflammatory Bowel Diseases. Int J Mol Sci 2021; 22:ijms222111682. [PMID: 34769112 PMCID: PMC8584226 DOI: 10.3390/ijms222111682] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel diseases (IBD) comprise a distinct set of clinical symptoms resulting from chronic inflammation within the gastrointestinal (GI) tract. Despite the significant progress in understanding the etiology and development of treatment strategies, IBD remain incurable for thousands of patients. Metabolic deregulation is indicative of IBD, including substantial shifts in lipid metabolism. Recent data showed that changes in some phospholipids are very common in IBD patients. For instance, phosphatidylcholine (PC)/phosphatidylethanolamine (PE) and lysophosphatidylcholine (LPC)/PC ratios are associated with the severity of the inflammatory process. Composition of phospholipids also changes upon IBD towards an increase in arachidonic acid and a decrease in linoleic and a-linolenic acid levels. Moreover, an increase in certain phospholipid metabolites, such as lysophosphatidylcholine, sphingosine-1-phosphate and ceramide, can result in enhanced intestinal inflammation, malignancy, apoptosis or necroptosis. Because some phospholipids are associated with pathogenesis of IBD, they may provide a basis for new strategies to treat IBD. Current attempts are aimed at controlling phospholipid and fatty acid levels through the diet or via pharmacological manipulation of lipid metabolism.
Collapse
|
15
|
|
16
|
Cortez‐Jugo C, Czuba‐Wojnilowicz E, Tan A, Caruso F. A Focus on "Bio" in Bio-Nanoscience: The Impact of Biological Factors on Nanomaterial Interactions. Adv Healthc Mater 2021; 10:e2100574. [PMID: 34170631 DOI: 10.1002/adhm.202100574] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/18/2021] [Indexed: 12/17/2022]
Abstract
Bio-nanoscience research encompasses studies on the interactions of nanomaterials with biological structures or what is commonly referred to as the biointerface. Fundamental studies on the influence of nanomaterial properties, including size, shape, composition, and charge, on the interaction with the biointerface have been central in bio-nanoscience to assess nanomaterial efficacy and safety for a range of biomedical applications. However, the state of the cells, tissues, or biological models can also influence the behavior of nanomaterials at the biointerface and their intracellular processing. Focusing on the "bio" in bio-nano, this review discusses the impact of biological properties at the cellular, tissue, and whole organism level that influences nanomaterial behavior, including cell type, cell cycle, tumor physiology, and disease states. Understanding how the biological factors can be addressed or exploited to enhance nanomaterial accumulation and uptake can guide the design of better and suitable models to improve the outcomes of materials in nanomedicine.
Collapse
Affiliation(s)
- Christina Cortez‐Jugo
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Ewa Czuba‐Wojnilowicz
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Abigail Tan
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|
17
|
Luo Z, Paunović N, Leroux JC. Physical methods for enhancing drug absorption from the gastrointestinal tract. Adv Drug Deliv Rev 2021; 175:113814. [PMID: 34052229 DOI: 10.1016/j.addr.2021.05.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 01/01/2023]
Abstract
Overcoming the gastrointestinal (GI) barriers is a formidable challenge in the oral delivery of active macromolecules such as peptide- and protein- based drugs. In the past four decades, a plethora of formulation strategies ranging from permeation enhancers, nanosized carriers, and chemical modifications of the drug's structure has been investigated to increase the oral absorption of these macromolecular compounds. However, only limited successes have been achieved so far, with the bioavailability of marketed oral peptide drugs remaining generally very low. Recently, a few approaches that are based on physical interactions, such as magnetic, acoustic, and mechanical forces, have been explored in order to control and improve the drug permeability across the GI mucosa. Although in the early stages, some of these methods have shown great potential both in terms of improved bioavailability and spatiotemporal delivery of drugs. Here, we offer a concise, yet critical overview of these rather unconventional technologies with a particular focus on their potential and possible challenges for further clinical translation.
Collapse
|
18
|
Emulsion-Based Multicompartment Vaginal Drug Carriers: From Nanoemulsions to Nanoemulgels. Int J Mol Sci 2021; 22:ijms22126455. [PMID: 34208652 PMCID: PMC8233730 DOI: 10.3390/ijms22126455] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 01/05/2023] Open
Abstract
In order to overcome the limitations associated with vaginal administration of drugs, e.g., the short contact time of the drug form with the mucosa or continuous carrier wash-out, the development of new carriers for gynecological use is necessary. Furthermore, high individual anatomical and physiological variability resulting in unsatisfactory therapeutic efficacy of lipophilic active substances requires application of multicompartment drug delivery systems. This manuscript provides an up-to-date comprehensive review of the literature on emulsion-based vaginal dosage forms (EVDF) including macroemulsions, microemulsions, nanoemulsions, multiple emulsions and self-emulsifying drug delivery systems. The first part of the paper discusses (i) the influence of anatomical-physiological conditions on therapeutic efficacy of drug forms after local and systemic administration, (ii) characterization of EVDF components and the manufacturing techniques of these dosage forms and (iii) methods used to evaluate the physicochemical and pharmaceutical properties of emulsion-based vaginal dosage forms. The second part of the paper presents (iv) the results of biological and in vivo studies as well as (v) clinical evaluation of EVDF safety and therapeutic efficacy across different indications.
Collapse
|
19
|
Wijesekara P, Liu Y, Wang W, Johnston EK, Sullivan MLG, Taylor RE, Ren X. Accessing and Assessing the Cell-Surface Glycocalyx Using DNA Origami. NANO LETTERS 2021; 21:4765-4773. [PMID: 34030445 PMCID: PMC8193633 DOI: 10.1021/acs.nanolett.1c01236] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/21/2021] [Indexed: 05/30/2023]
Abstract
The cell-surface glycocalyx serves as a physiological barrier regulating cellular accessibility to macromolecules and other cells. Conventional glycocalyx characterization has largely been morphological rather than functional. Here, we demonstrated direct glycocalyx anchoring of DNA origami nanotiles and performed a comprehensive comparison with traditional origami targeting to the phospholipid bilayer (PLB) using cholesterol. While DNA nanotiles effectively accessed single-stranded DNA initiators anchored on the glycocalyx, their accessibility to the underlying PLB was only permitted by extended nanotile-to-initiator spacing or by enzymatic glycocalyx degradation using trypsin or pathogenic neuraminidase. Thus, the DNA nanotiles, being expelled by the physiologic glycocalyx, provide an effective functional measure of the glycocalyx barrier integrity and faithfully predict cell-to-cell accessibility during DNA-guided multicellular assembly. Lastly, the glycocalyx-anchoring mechanism enabled enhanced cell-surface stability and cellular uptake of nanotiles compared to PLB anchoring. This research lays the foundation for future development of DNA nanodevices to access the cell surface.
Collapse
Affiliation(s)
- Piyumi Wijesekara
- Department
of Biomedical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Ying Liu
- Department
of Mechanical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Weitao Wang
- Department
of Mechanical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Elizabeth K. Johnston
- Department
of Biomedical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Mara L. G. Sullivan
- Center
for Biologic Imaging, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, Pennsylvania, United States
| | - Rebecca E. Taylor
- Department
of Biomedical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
- Department
of Mechanical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
- Department
of Electrical and Computer Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Xi Ren
- Department
of Biomedical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
- Department
of Mechanical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
20
|
Wagner C, Torow N, Hornef MW, Lelouard H. Spatial and temporal key steps in early-life intestinal immune system development and education. FEBS J 2021; 289:4731-4757. [PMID: 34076962 DOI: 10.1111/febs.16047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/15/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
Education of our intestinal immune system early in life strongly influences adult health. This education strongly relies on series of events that must occur in well-defined time windows. From initial colonization by maternal-derived microbiota during delivery to dietary changes from mother's milk to solid foods at weaning, these early-life events have indeed long-standing consequences on our immunity, facilitating tolerance to environmental exposures or, on the contrary, increasing the risk of developing noncommunicable diseases such as allergies, asthma, obesity, and inflammatory bowel diseases. In this review, we provide an outline of the recent advances in our understanding of these events and how they are mechanistically related to intestinal immunity development and education. First, we review the susceptibility of neonates to infections and inflammatory diseases, related to their immune system and microbiota changes. Then, we highlight the maternal factors involved in protection and education of the mucosal immune system of the offspring, the role of the microbiota, and the nature of neonatal immune system until weaning. We also present how the development of some immune responses is intertwined in temporal and spatial windows of opportunity. Finally, we discuss pending questions regarding the neonate particular immune status and the activation of the intestinal immune system at weaning.
Collapse
Affiliation(s)
- Camille Wagner
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Natalia Torow
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | | |
Collapse
|
21
|
Qin Q, Lang S, Huang X. Synthetic linear glycopolymers and their biological applications. J Carbohydr Chem 2021; 40:1-44. [PMID: 35308080 PMCID: PMC8932951 DOI: 10.1080/07328303.2021.1928156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/06/2021] [Indexed: 10/21/2022]
Abstract
As typical affinities of carbohydrates with their receptors are modest, polymers of carbohydrates (glycopolymers) are exciting tools to probe the multifaceted biological activities of glycans. In this review, the linear glycopolymers and the multivalency effects are first introduced. This is followed by discussions of methods to synthesize these polymers. Subsequently, the interactions of glycopolymers with plant lectins and viral/bacterial carbohydrate binding proteins are discussed. In addition, applications of the glycopolymers in facilitating glycan microarray studies, mimicking cell surface glycans, modulation of the immune system, cryoprotection of protein, and electron-beam lithography are presented to stimulate further development of this fascinating technology.
Collapse
Affiliation(s)
- Qian Qin
- Department of Chemistry, Michigan StateUniversity, East Lansing, MI, USA
| | - Shuyao Lang
- Department of Chemistry, Michigan StateUniversity, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan StateUniversity, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
22
|
Fedi A, Vitale C, Ponschin G, Ayehunie S, Fato M, Scaglione S. In vitro models replicating the human intestinal epithelium for absorption and metabolism studies: A systematic review. J Control Release 2021; 335:247-268. [PMID: 34033859 DOI: 10.1016/j.jconrel.2021.05.028] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/17/2022]
Abstract
Absorption, distribution, metabolism and excretion (ADME) studies represent a fundamental step in the early stages of drug discovery. In particular, the absorption of orally administered drugs, which occurs at the intestinal level, has gained attention since poor oral bioavailability often led to failures for new drug approval. In this context, several in vitro preclinical models have been recently developed and optimized to better resemble human physiology in the lab and serve as an animal alternative to accomplish the 3Rs principles. However, numerous models are ineffective in recapitulating the key features of the human small intestine epithelium and lack of prediction potential for drug absorption and metabolism during the preclinical stage. In this review, we provide an overview of in vitro models aimed at mimicking the intestinal barrier for pharmaceutical screening. After briefly describing how the human small intestine works, we present i) conventional 2D synthetic and cell-based systems, ii) 3D models replicating the main features of the intestinal architecture, iii) micro-physiological systems (MPSs) reproducing the dynamic stimuli to which cells are exposed in the native microenvironment. In this review, we will highlight the benefits and drawbacks of the leading intestinal models used for drug absorption and metabolism studies.
Collapse
Affiliation(s)
- Arianna Fedi
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy; National Research Council of Italy, Institute of Electronics, Computer and Telecommunications (IEIIT) Institute, 16149 Genoa, Italy
| | - Chiara Vitale
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunications (IEIIT) Institute, 16149 Genoa, Italy
| | - Giulia Ponschin
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
| | | | - Marco Fato
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy; National Research Council of Italy, Institute of Electronics, Computer and Telecommunications (IEIIT) Institute, 16149 Genoa, Italy
| | - Silvia Scaglione
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunications (IEIIT) Institute, 16149 Genoa, Italy.
| |
Collapse
|
23
|
Arroyo Portilla C, Tomas J, Gorvel JP, Lelouard H. From Species to Regional and Local Specialization of Intestinal Macrophages. Front Cell Dev Biol 2021; 8:624213. [PMID: 33681185 PMCID: PMC7930007 DOI: 10.3389/fcell.2020.624213] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Initially intended for nutrient uptake, phagocytosis represents a central mechanism of debris removal and host defense against invading pathogens through the entire animal kingdom. In vertebrates and also many invertebrates, macrophages (MFs) and MF-like cells (e.g., coelomocytes and hemocytes) are professional phagocytic cells that seed tissues to maintain homeostasis through pathogen killing, efferocytosis and tissue shaping, repair, and remodeling. Some MF functions are common to all species and tissues, whereas others are specific to their homing tissue. Indeed, shaped by their microenvironment, MFs become adapted to perform particular functions, highlighting their great plasticity and giving rise to high population diversity. Interestingly, the gut displays several anatomic and functional compartments with large pools of strikingly diversified MF populations. This review focuses on recent advances on intestinal MFs in several species, which have allowed to infer their specificity and functions.
Collapse
Affiliation(s)
- Cynthia Arroyo Portilla
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Departamento de Análisis Clínicos, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Julie Tomas
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | |
Collapse
|
24
|
das Neves J, Sverdlov Arzi R, Sosnik A. Molecular and cellular cues governing nanomaterial-mucosae interactions: from nanomedicine to nanotoxicology. Chem Soc Rev 2021; 49:5058-5100. [PMID: 32538405 DOI: 10.1039/c8cs00948a] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mucosal tissues constitute the largest interface between the body and the surrounding environment and they regulate the access of molecules, supramolecular structures, particulate matter, and pathogens into it. All mucosae are characterized by an outer mucus layer that protects the underlying cells from physicochemical, biological and mechanical insults, a mono-layered or stratified epithelium that forms tight junctions and controls the selective transport of solutes across it and associated lymphoid tissues that play a sentinel role. Mucus is a gel-like material comprised mainly of the glycoprotein mucin and water and it displays both hydrophilic and hydrophobic domains, a net negative charge, and high porosity and pore interconnectivity, providing an efficient barrier for the absorption of therapeutic agents. To prolong the residence time, absorption and bioavailability of a broad spectrum of active compounds upon mucosal administration, mucus-penetrating and mucoadhesive particles have been designed by tuning the chemical composition, the size, the density, and the surface properties. The benefits of utilizing nanomaterials that interact intimately with mucosae by different mechanisms in the nanomedicine field have been extensively reported. To ensure the safety of these nanosystems, their compatibility is evaluated in vitro and in vivo in preclinical and clinical trials. Conversely, there is a growing concern about the toxicity of nanomaterials dispersed in air and water effluents that unintentionally come into contact with the airways and the gastrointestinal tract. Thus, deep understanding of the key nanomaterial properties that govern the interplay with mucus and tissues is crucial for the rational design of more efficient drug delivery nanosystems (nanomedicine) and to anticipate the fate and side-effects of nanoparticulate matter upon acute or chronic exposure (nanotoxicology). This review initially overviews the complex structural features of mucosal tissues, including the structure of mucus, the epithelial barrier, the mucosal-associated lymphatic tissues and microbiota. Then, the most relevant investigations attempting to identify and validate the key particle features that govern nanomaterial-mucosa interactions and that are relevant in both nanomedicine and nanotoxicology are discussed in a holistic manner. Finally, the most popular experimental techniques and the incipient use of mathematical and computational models to characterize these interactions are described.
Collapse
Affiliation(s)
- José das Neves
- i3S - Instituto de Investigação e Inovação em Saúde & INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Roni Sverdlov Arzi
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Haifa, 3200003, Israel.
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Haifa, 3200003, Israel.
| |
Collapse
|
25
|
Bjørgen H, Li Y, Kortner TM, Krogdahl Å, Koppang EO. Anatomy, immunology, digestive physiology and microbiota of the salmonid intestine: Knowns and unknowns under the impact of an expanding industrialized production. FISH & SHELLFISH IMMUNOLOGY 2020; 107:172-186. [PMID: 32979510 DOI: 10.1016/j.fsi.2020.09.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
Increased industrialized production of salmonids challenges aspects concerning available feed resources and animal welfare. The immune system plays a key component in this respect. Novel feed ingredients may trigger unwarranted immune responses again affecting the well-being of the fish. Here we review our current knowledge concerning salmon intestinal anatomy, immunity, digestive physiology and microbiota in the context of industrialized feeding regimes. We point out knowledge gaps and indicate promising novel technologies to improve salmonid intestinal health.
Collapse
Affiliation(s)
- Håvard Bjørgen
- Section of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Yanxian Li
- Nutrition and Health Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Trond M Kortner
- Nutrition and Health Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Åshild Krogdahl
- Nutrition and Health Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Erling Olaf Koppang
- Section of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| |
Collapse
|
26
|
Abstract
In the current era, many formulations have been designed in the form of vesicular carriers
like liposomes and niosomes which have been proved to be one of the potential candidates for
drug delivery by the oral route but due to the gastrointestinal environment i.e. pH, presence of enzymes,
and bile salts, their use is limited. Because of these difficulties, research is being done to
increase the stability and efficacy of the drug. Thus bilosomes have been developed as a potential
vesicular carrier system for oral vaccine delivery, transdermal and parenteral targeted drug delivery.
The present article covers various aspects related to the novel vesicular system that is based on bile
salts called bilosomes, for targetted drug delivery systems. It includes information related to bilosome
composition, formulation techniques, characterization methods, applications in oral immunization
as vaccine delivery approach and advantages over conventional nanocarriers such as
liposomes and niosomes. It also focuses on the stability and applications of bilosomes along
with scalability and potentiality in biomedical field of oral immunization against various dreadful
diseases.
Collapse
Affiliation(s)
- Pradnya Palekar-Shanbhag
- Department of Pharmaceutics, Oriental College of Pharmacy, Sanpada, University of Mumbai, Mumbai, India
| | - Supriya Lande
- Department of Pharmaceutics, Oriental College of Pharmacy, Sanpada, University of Mumbai, Mumbai, India
| | - Riya Chandra
- Department of Pharmaceutics, Oriental College of Pharmacy, Sanpada, University of Mumbai, Mumbai, India
| | - Drushti Rane
- Department of Pharmaceutics, Oriental College of Pharmacy, Sanpada, University of Mumbai, Mumbai, India
| |
Collapse
|
27
|
Piotrowska M, Swierczynski M, Fichna J, Piechota-Polanczyk A. The Nrf2 in the pathophysiology of the intestine: Molecular mechanisms and therapeutic implications for inflammatory bowel diseases. Pharmacol Res 2020; 163:105243. [PMID: 33080322 DOI: 10.1016/j.phrs.2020.105243] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/29/2020] [Accepted: 10/04/2020] [Indexed: 12/14/2022]
Abstract
Nrf2 (nuclear factor erythroid 2-related factor 2) is a stress-responsive transcription factor, associated with cellular homeostasis. Under normal conditions Nrf2 is kept in the cytoplasm by Kelch-like ECH-associated protein 1 (Keap1) which facilitates its degradation. Meanwhile, oxidative or electrophilic stress trigger Keap1 dissociation from the Nrf2/Keap1 complex and Nrf2 translocation to the nucleus where it induces the expression of numerous anti-oxidative and anti-inflammatory genes. The Nrf2/Keap1 axis plays a crucial role in the development of gastrointestinal (GI) tract and the maintenance of its proper functionality. This axis also seems to be a promising candidate for prevention of inflammatory bowel diseases (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), as well as their severe complications such as intestinal fibrosis and colorectal cancer. This review focuses on the role of Nrf2/Keap1 in 1) the development and proper functionality of GI tract, 2) the pathophysiology of GI diseases and their long-term complications, 3) the effectiveness of currently used drugs and non-conventional treatments which influence Nrf2/Keap1 and are potentially effective in IBD treatment, as well as 4) the effect of gut microbiota on Nrf2/Keap1 pathway in IBD.
Collapse
Affiliation(s)
- Marta Piotrowska
- Department of Biochemistry, Faculty of Medicine, Medical UniverSity of Lodz, Poland
| | - Mikolaj Swierczynski
- Department of Biochemistry, Faculty of Medicine, Medical UniverSity of Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical UniverSity of Lodz, Poland
| | - Aleksandra Piechota-Polanczyk
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow 30-387, Poland.
| |
Collapse
|
28
|
Sun S, Jiang S, Wang J, Chen C, Han S, Che H. Cholera toxin induces food allergy through Th2 cell differentiation which is unaffected by Jagged2. Life Sci 2020; 263:118514. [PMID: 33010283 DOI: 10.1016/j.lfs.2020.118514] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 01/05/2023]
Abstract
AIMS Cholera toxin is often used to induce food allergies. However, its exact mode of action and effect remain ambiguous. In this study, we established a BALB/c mouse cholera toxin/ovalbumin-induced food allergy model to determine the molecular basis and signaling mechanisms of the immune regulation of cholera toxin during food allergy. MATERIALS AND METHODS The adjuvant activity of cholera toxin was analyzed by establishing mouse allergy model, and the allergic reaction of each group of mice was evaluated. The effect of cholera toxin on Th1/Th2 cell differentiation was analyzed to further explore the role of cholera toxin in allergen immune response. We stimulated bone marrow-derived dendritic cells (BMDCs) with cholera toxin in vitro to investigate the effect of cholera toxin on Notch ligand expression. BMDCs and naive CD4+T cells were co-cultured in vitro, and their cytokine levels were examined to investigate whether cholera toxin regulates Th cell differentiation via the Jagged2 Notch signaling pathway. KEY FINDINGS The results showed that in the presence of allergens, cholera toxin promotes Th2 cell differentiation and enhances the body's immune response. Cholera toxin induces expression of the Notch ligand Jagged2, but Jagged2 Notch signaling pathway is not required to promote BMDCs-mediated differentiation of Th2 cells. SIGNIFICANCE This study initially revealed the mechanism by which cholera toxin plays an adjuvant role in food allergy, and provides reference for future related research.
Collapse
Affiliation(s)
- Shanfeng Sun
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Songsong Jiang
- College of Food Science and Engineering, Yangzhou University, No.88 Daxue South Road, Hanjiang District, Yangzhou, Jiangsu Province, China
| | - Junjuan Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Cheng Chen
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Shiwen Han
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Huilian Che
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
29
|
Kudelka MR, Stowell SR, Cummings RD, Neish AS. Intestinal epithelial glycosylation in homeostasis and gut microbiota interactions in IBD. Nat Rev Gastroenterol Hepatol 2020; 17:597-617. [PMID: 32710014 PMCID: PMC8211394 DOI: 10.1038/s41575-020-0331-7] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/05/2020] [Indexed: 02/08/2023]
Abstract
Inflammatory bowel disease (IBD) affects 6.8 million people globally. A variety of factors have been implicated in IBD pathogenesis, including host genetics, immune dysregulation and gut microbiota alterations. Emerging evidence implicates intestinal epithelial glycosylation as an underappreciated process that interfaces with these three factors. IBD is associated with increased expression of truncated O-glycans as well as altered expression of terminal glycan structures. IBD genes, glycosyltransferase mislocalization, altered glycosyltransferase and glycosidase expression and dysbiosis drive changes in the glycome. These glycan changes disrupt the mucus layer, glycan-lectin interactions, host-microorganism interactions and mucosal immunity, and ultimately contribute to IBD pathogenesis. Epithelial glycans are especially critical in regulating the gut microbiota through providing bacterial ligands and nutrients and ultimately determining the spatial organization of the gut microbiota. In this Review, we discuss the regulation of intestinal epithelial glycosylation, altered epithelial glycosylation in IBD and mechanisms for how these alterations contribute to disease pathobiology. We hope that this Review provides a foundation for future studies on IBD glycosylation and the emergence of glycan-inspired therapies for IBD.
Collapse
Affiliation(s)
- Matthew R Kudelka
- Medical Scientist Training Program, Emory University School of Medicine, Atlanta, GA, USA
- Department of Internal Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sean R Stowell
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
30
|
Sotomayor-Gerding D, Troncoso JM, Pino A, Almendras F, Diaz MR. Assessing the Immune Response of Atlantic Salmon ( Salmo salar) after the Oral Intake of Alginate-Encapsulated Piscirickettsia salmonis Antigens. Vaccines (Basel) 2020; 8:vaccines8030450. [PMID: 32796725 PMCID: PMC7565443 DOI: 10.3390/vaccines8030450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022] Open
Abstract
Salmon rickettsial septicaemia (SRS) is the infectious disease that produces the highest losses in the Chilean salmon industry. As a new strategy for the control of SRS outbreaks, in this study we evaluated the effect of alginate-encapsulated Piscirickettsia salmonis antigens (AEPSA) incorporated in the feed as an oral vaccine to induce the immune response in Atlantic salmon (Salmo salar). Fish were distributed into three vaccination groups (injectable, oral high dose, oral low dose). Feed intake and fish growth were recorded during the trial. The P. salmonis-specific IgM levels in blood plasma were measured by ELISA. Alginate microparticles containing the antigen were effectively incorporated in fish feed to produce the oral vaccine. Incorporation of AEPSA did not affect the palatability of the feed or the fish appetite. Furthermore, the oral vaccine did not have a negative effect on fish growth. Finally, the oral vaccine (high and low dose) produced an acquired immune response (IgM) similar to the injectable vaccine, generating a statistically significant increase in the IgM levels at 840-degree days for both experimental groups. These findings suggest that AEPSA incorporated in the feed can be an effective alternative to boost the immune response in Atlantic salmon (S. salar).
Collapse
Affiliation(s)
- Daniela Sotomayor-Gerding
- Programa de Doctorado en Ciencias de Recursos Naturales, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Correspondence: (D.S.-G.); (M.R.D.); Tel.: +56-45-2744240 (D.S.-G.); +56-45-2744232 (M.R.D.)
| | | | - Alejandro Pino
- Anasac Chile S.A., Veterinary Division, Almirante Pastene 300, Providencia 7500534, Santiago, Chile;
| | | | - Mónica Rubilar Diaz
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Scientific and Technological Bioresource Nucleus, BIOREN, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Correspondence: (D.S.-G.); (M.R.D.); Tel.: +56-45-2744240 (D.S.-G.); +56-45-2744232 (M.R.D.)
| |
Collapse
|
31
|
Chaikhumwang P, Kitsongsermthon J, Manopakdee K, Chongcharoen W, Nilubol D, Chanvorachote P, Somparn P, Tantituvanont A. Cationic Polylactic Acid-Based Nanoparticles Improve BSA-FITC Transport Across M Cells and Engulfment by Porcine Alveolar Macrophages. AAPS PharmSciTech 2020; 21:134. [PMID: 32415347 DOI: 10.1208/s12249-020-01689-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/13/2020] [Indexed: 01/14/2023] Open
Abstract
This work described the development of a cationic polylactic acid (PLA)-based nanoparticles (NPs) as an antigen delivery system using dimethyldioctadecylammonium bromide (DDA) to facilitate the engulfment of BSA-FITC by porcine alveolar macrophages (3D4/2 cells) and heat-labile enterotoxin subunit B (LTB) to enhance the transport of BSA-FITC across M cells. The experimental design methodology was employed to study the influence of PLA, polyvinyl alcohol (PVA), DDA, and LTB on the physical properties of the PLA-based NPs. The size of selected cationic PLA NPs comprising 5% PLA, 5% PVA, and 0.6% DDA with or without LTB absorption was range from 367 to 390 nm with a polydispersity index of 0.26, a zeta potential of + 26.00 to + 30.55 mV, and entrapment efficiency of 41.43%. Electron micrographs revealed NPs with spherical shape. The release kinetic of BSA from the NPs followed the Korsmeyer-Peppas kinetics. The cationic PLA NPs with LTB surface absorption showed 3-fold increase in BSA-FITC transported across M cells compared with the NPs without LTB absorption. The uptake studies demonstrated 2-fold increase in BSA-FITC intensity in 3D4/2 cells with cationic NPs as compared with anionic NPs. Overall, the results suggested that LTB decreased the retention time of BSA-FITC loaded in the cationic PLA NPs within the M cells, thus promoting the transport of BSA-FITC across the M cells, and cationic NPs composed of DDA help facilitate the uptake of BSA-FITC in the 3D4/2 cells. Further studies in pigs with respiratory antigens will provide information on the efficacy of cationic PLA NPs as a nasal antigen carrier system.
Collapse
|
32
|
Hua S. Advances in Oral Drug Delivery for Regional Targeting in the Gastrointestinal Tract - Influence of Physiological, Pathophysiological and Pharmaceutical Factors. Front Pharmacol 2020; 11:524. [PMID: 32425781 PMCID: PMC7212533 DOI: 10.3389/fphar.2020.00524] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 04/03/2020] [Indexed: 12/22/2022] Open
Abstract
The oral route is by far the most common route of drug administration in the gastrointestinal tract and can be used for both systemic drug delivery and for treating local gastrointestinal diseases. It is the most preferred route by patients, due to its advantages, such as ease of use, non-invasiveness, and convenience for self-administration. Formulations can also be designed to enhance drug delivery to specific regions in the upper or lower gastrointestinal tract. Despite the clear advantages offered by the oral route, drug delivery can be challenging as the human gastrointestinal tract is complex and displays a number of physiological barriers that affect drug delivery. Among these challenges are poor drug stability, poor drug solubility, and low drug permeability across the mucosal barriers. Attempts to overcome these issues have focused on improved understanding of the physiology of the gastrointestinal tract in both healthy and diseased states. Innovative pharmaceutical approaches have also been explored to improve regional drug targeting in the gastrointestinal tract, including nanoparticulate formulations. This review will discuss the physiological, pathophysiological, and pharmaceutical considerations influencing drug delivery for the oral route of administration, as well as the conventional and novel drug delivery approaches. The translational challenges and development aspects of novel formulations will also be addressed.
Collapse
Affiliation(s)
- Susan Hua
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
33
|
Frey A, Lunding LP, Ehlers JC, Weckmann M, Zissler UM, Wegmann M. More Than Just a Barrier: The Immune Functions of the Airway Epithelium in Asthma Pathogenesis. Front Immunol 2020; 11:761. [PMID: 32411147 PMCID: PMC7198799 DOI: 10.3389/fimmu.2020.00761] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/03/2020] [Indexed: 12/11/2022] Open
Abstract
Allergic bronchial asthma is a chronic disease of the airways that is characterized by symptoms like respiratory distress, chest tightness, wheezing, productive cough, and acute episodes of broncho-obstruction. This symptom-complex arises on the basis of chronic allergic inflammation of the airway wall. Consequently, the airway epithelium is central to the pathogenesis of this disease, because its multiple abilities directly have an impact on the inflammatory response and thus the formation of the disease. In turn, its structure and functions are markedly impaired by the inflammation. Hence, the airway epithelium represents a sealed, self-cleaning barrier, that prohibits penetration of inhaled allergens, pathogens, and other noxious agents into the body. This barrier is covered with mucus that further contains antimicrobial peptides and antibodies that are either produced or specifically transported by the airway epithelium in order to trap these particles and to remove them from the body by a process called mucociliary clearance. Once this first line of defense of the lung is overcome, airway epithelial cells are the first cells to get in contact with pathogens, to be damaged or infected. Therefore, these cells release a plethora of chemokines and cytokines that not only induce an acute inflammatory reaction but also have an impact on the alignment of the following immune reaction. In case of asthma, all these functions are impaired by the already existing allergic immune response that per se weakens the barrier integrity and self-cleaning abilities of the airway epithelium making it more vulnerable to penetration of allergens as well as of infection by bacteria and viruses. Recent studies indicate that the history of allergy- and pathogen-derived insults can leave some kind of memory in these cells that can be described as imprinting or trained immunity. Thus, the airway epithelium is in the center of processes that lead to formation, progression and acute exacerbation of asthma.
Collapse
Affiliation(s)
- Andreas Frey
- Division of Mucosal Immunology and Diagnostics, Research Center Borstel, Borstel, Germany.,Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany
| | - Lars P Lunding
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Division of Asthma Exacerbation & Regulation, Research Center Borstel, Borstel, Germany
| | - Johanna C Ehlers
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Division of Experimental Pneumology, Research Center Borstel, Borstel, Germany
| | - Markus Weckmann
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Department of Pediatric Pulmonology and Allergology, University Children's Hospital, Lübeck, Germany
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany.,Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Michael Wegmann
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Division of Asthma Exacerbation & Regulation, Research Center Borstel, Borstel, Germany
| |
Collapse
|
34
|
Homayun B, Choi HJ. Halloysite nanotube-embedded microparticles for intestine-targeted co-delivery of biopharmaceuticals. Int J Pharm 2020; 579:119152. [DOI: 10.1016/j.ijpharm.2020.119152] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/29/2020] [Accepted: 02/09/2020] [Indexed: 12/11/2022]
|
35
|
Liposomes for Enhanced Bioavailability of Water-Insoluble Drugs: In Vivo Evidence and Recent Approaches. Pharmaceutics 2020; 12:pharmaceutics12030264. [PMID: 32183185 PMCID: PMC7151102 DOI: 10.3390/pharmaceutics12030264] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 12/26/2022] Open
Abstract
It has been known that a considerable number of drugs in clinical use or under development are water-insoluble drugs with poor bioavailability (BA). The liposomal delivery system has drawn attention as one of the noteworthy approaches to increase dissolution and subsequently absorption in the gastrointestinal (GI) tract because of its biocompatibility and ability to encapsulate hydrophobic molecules in the lipid domain. However, there have been several drawbacks, such as structural instability in the GI tract and poor permeability across intestinal epithelia because of its relatively large size. In addition, there have been no liposomal formulations approved for oral use to date, despite the success of parenteral liposomes. Nevertheless, liposomal oral delivery has resurged with the rapid increase of published studies in the last decade. However, it is discouraging that most of this research has been in vitro studies only and there have not been many water-insoluble drugs with in vivo data. The present review focused on the in vivo evidence for the improved BA of water-insoluble drugs using liposomes to resolve doubts raised concerning liposomal oral delivery and attempted to provide insight by highlighting the approaches used for in vivo achievements.
Collapse
|
36
|
Sun WW, Krystofiak ES, Leo-Macias A, Cui R, Sesso A, Weigert R, Ebrahim S, Kachar B. Nanoarchitecture and dynamics of the mouse enteric glycocalyx examined by freeze-etching electron tomography and intravital microscopy. Commun Biol 2020; 3:5. [PMID: 31925335 PMCID: PMC6946683 DOI: 10.1038/s42003-019-0735-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022] Open
Abstract
The glycocalyx is a highly hydrated, glycoprotein-rich coat shrouding many eukaryotic and prokaryotic cells. The intestinal epithelial glycocalyx, comprising glycosylated transmembrane mucins, is part of the primary host-microbe interface and is essential for nutrient absorption. Its disruption has been implicated in numerous gastrointestinal diseases. Yet, due to challenges in preserving and visualizing its native organization, glycocalyx structure-function relationships remain unclear. Here, we characterize the nanoarchitecture of the murine enteric glycocalyx using freeze-etching and electron tomography. Micrometer-long mucin filaments emerge from microvillar-tips and, through zigzagged lateral interactions form a three-dimensional columnar network with a 30 nm mesh. Filament-termini converge into globular structures ~30 nm apart that are liquid-crystalline packed within a single plane. Finally, we assess glycocalyx deformability and porosity using intravital microscopy. We argue that the columnar network architecture and the liquid-crystalline packing of the filament termini allow the glycocalyx to function as a deformable size-exclusion filter of luminal contents. Sun, Krystofiak et al. show the nanoarchitecture of the murine enteric glycocalyx, glycoprotein-rich coat covering cells and assess its porosity and deformability in mice, providing a comprehensive structural framework. This study suggests that the glycocalyx may function as a deformable size-exclusion filter of luminal contents.
Collapse
Affiliation(s)
- Willy W Sun
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA.,Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD, 20740, USA
| | - Evan S Krystofiak
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alejandra Leo-Macias
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Runjia Cui
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Antonio Sesso
- Sector of Structural Biology, Institute of Tropical Medicine, University of São Paulo, Sao Paulo, SP, 05403, Brazil
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Seham Ebrahim
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
37
|
Glyco-nanoparticles: New drug delivery systems in cancer therapy. Semin Cancer Biol 2019; 69:24-42. [PMID: 31870939 DOI: 10.1016/j.semcancer.2019.12.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/28/2019] [Accepted: 12/02/2019] [Indexed: 12/24/2022]
Abstract
Cancer is known as one of the most common diseases that are associated with high mobility and mortality in the world. Despite several efforts, current cancer treatment modalities often are highly toxic and lack efficacy and specificity. However, the application of nanotechnology has led to the development of effective nanosized drug delivery systems which are highly selective for tumors and allow a slow release of active anticancer agents. Different Nanoparticles (NPs) such as the silicon-based nano-materials, polymers, liposomes and metal NPs have been designed to deliver anti-cancer drugs to tumor sites. Among different drug delivery systems, carbohydrate-functionalized nanomaterials, specially based on their multi-valent binding capacities and desirable bio-compatibility, have attracted considerable attention as an excellent candidate for controlled release of therapeutic agents. In addition, these carbohydrate functionalized nano-carriers are more compatible with construction of the intracellular delivery platforms like the carbohydrate-modified metal NPs, quantum dots, and magnetic nano-materials. In this review, we discuss recent research in the field of multifunctional glycol-nanoparticles (GNPs) intended for cancer drug delivery applications.
Collapse
|
38
|
O’Callaghan AA, Corr SC. Establishing Boundaries: The Relationship That Exists between Intestinal Epithelial Cells and Gut-Dwelling Bacteria. Microorganisms 2019; 7:microorganisms7120663. [PMID: 31818022 PMCID: PMC6956261 DOI: 10.3390/microorganisms7120663] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/05/2019] [Accepted: 12/07/2019] [Indexed: 12/16/2022] Open
Abstract
The human gastrointestinal (GI) tract is a highly complex organ in which various dynamic physiological processes are tightly coordinated while interacting with a complex community of microorganisms. Within the GI tract, intestinal epithelial cells (IECs) create a structural interface that separates the intestinal lumen from the underlying lamina propria. In the lumen, gut-dwelling microbes play an essential role in maintaining gut homeostasis and functionality. Whether commensal or pathogenic, their interaction with IECs is inevitable. IECs and myeloid immune cells express an array of pathogen recognition receptors (PRRs) that define the interaction of both pathogenic and beneficial bacteria with the intestinal mucosa and mount appropriate responses including induction of barrier-related factors which enhance the integrity of the epithelial barrier. Indeed, the integrity of this barrier and induction of appropriate immune responses is critical to health status, with defects in this barrier and over-activation of immune cells by invading microbes contributing to development of a range of inflammatory and infectious diseases. This review describes the complexity of the GI tract and its interactions with gut bacteria.
Collapse
|
39
|
A Bioinformatics View of Glycan⁻Virus Interactions. Viruses 2019; 11:v11040374. [PMID: 31018588 PMCID: PMC6521074 DOI: 10.3390/v11040374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023] Open
Abstract
Evidence of the mediation of glycan molecules in the interaction between viruses and their hosts is accumulating and is now partially reflected in several online databases. Bioinformatics provides convenient and efficient means of searching, visualizing, comparing, and sometimes predicting, interactions in numerous and diverse molecular biology applications related to the -omics fields. As viromics is gaining momentum, bioinformatics support is increasingly needed. We propose a survey of the current resources for searching, visualizing, comparing, and possibly predicting host–virus interactions that integrate the presence and role of glycans. To the best of our knowledge, we have mapped the specialized and general-purpose databases with the appropriate focus. With an illustration of their potential usage, we also discuss the strong and weak points of the current bioinformatics landscape in the context of understanding viral infection and the immune response to it.
Collapse
|
40
|
Evaluating Shigella flexneri Pathogenesis in the Human Enteroid Model. Infect Immun 2019; 87:IAI.00740-18. [PMID: 30642900 PMCID: PMC6434113 DOI: 10.1128/iai.00740-18] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/06/2019] [Indexed: 01/02/2023] Open
Abstract
The enteric pathogen Shigella is one of the leading causes of moderate-to-severe diarrhea and death in young children in developing countries. Transformed cell lines and animal models have been widely used to study Shigella pathogenesis. The enteric pathogen Shigella is one of the leading causes of moderate-to-severe diarrhea and death in young children in developing countries. Transformed cell lines and animal models have been widely used to study Shigella pathogenesis. In addition to altered physiology, transformed cell lines are composed of a single cell type that does not sufficiently represent the complex multicellular environment of the human colon. Most available animal models do not accurately mimic human disease. The human intestinal enteroid model, derived from LGR5+ stem cell-containing intestinal crypts from healthy subjects, represents a technological leap in human gastrointestinal system modeling and provides a more physiologically relevant system that includes multiple cell types and features of the human intestine. We established the utility of this model for studying basic aspects of Shigella pathogenesis and host responses. In this study, we show that Shigellaflexneri is capable of infecting and replicating intracellularly in human enteroids derived from different segments of the intestine. Apical invasion by S. flexneri is very limited but increases ∼10-fold when enteroids are differentiated to include M cells. Invasion via the basolateral surface was at least 2-log10 units more efficient than apical infection. Increased secretion of interleukin-8 and higher expression levels of the mucin glycoprotein Muc2 were observed in the enteroids following S. flexneri infection. The human enteroid model promises to bridge some of the gaps between traditional cell culture, animal models, and human infection.
Collapse
|
41
|
Frey A, Ramaker K, Röckendorf N, Wollenberg B, Lautenschläger I, Gébel G, Giemsa A, Heine M, Bargheer D, Nielsen P. Fate and Translocation of (Nano)Particulate Matter in the Gastrointestinal Tract. BIOLOGICAL RESPONSES TO NANOSCALE PARTICLES 2019. [DOI: 10.1007/978-3-030-12461-8_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
42
|
Melkoumov A, St-Jean I, Banquy X, Leclair G, Leblond Chain J. GM1-Binding Conjugates To Improve Intestinal Permeability. Mol Pharm 2018; 16:60-70. [PMID: 30422668 DOI: 10.1021/acs.molpharmaceut.8b00776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Drugs and proteins with poor intestinal permeability have a limited oral bioavailability. To remediate this problem, a receptor-mediated endocytosis and transcytosis approach was explored. Indeed, the nontoxic β subunit of cholera toxin (CTB) can cross the intestinal barrier by binding to receptor GM1. In this study, we explored the use of GM1-binding peptides and CTB as potential covalent carriers of poorly permeable molecules. GM1-binding peptides (G23, P3) and CTB were conjugated to poorly permeable fluorescent probes such as fluorescein isothiocyanate (FITC) and albumin-FITC using triethylene glycol spacers and click chemistry. The affinity of the peptide conjugates with receptor GM1 was confirmed by isothermal titration calorimetry or microscale thermophoresis, and the results suggested the involvement of nonspecific interactions. Conjugating the model drugs to G23 and P3 improved the internalization into Caco-2 and T84 cells, although the process was not dependent on the amount of GM1 receptor. However, conjugation of bovine serum albumin FITC to CTB increased the internalization in the same cells in a GM1-dependent pathway. Peptide conjugates demonstrated a limited permeability through a Caco-2 monolayer, whereas G23 and CTB conjugates slightly enhanced permeability through a T84 cell monolayer compared to model drugs alone. Since CTB can improve the permeability of large macromolecules such as albumin, it is an interesting carrier for the improvement of oral bioavailability of various other macromolecules such as heparins, proteins, and siRNAs.
Collapse
Affiliation(s)
- Alexandre Melkoumov
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| | - Isabelle St-Jean
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| | - Xavier Banquy
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| | - Grégoire Leclair
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| | - Jeanne Leblond Chain
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| |
Collapse
|
43
|
Reinholz J, Landfester K, Mailänder V. The challenges of oral drug delivery via nanocarriers. Drug Deliv 2018; 25:1694-1705. [PMID: 30394120 PMCID: PMC6225504 DOI: 10.1080/10717544.2018.1501119] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/06/2018] [Accepted: 07/12/2018] [Indexed: 01/11/2023] Open
Abstract
The oral application of pharmaceuticals is unarguably the most convenient method of application. Especially for protein- or peptide-based drugs, however, the effectiveness is significantly reduced due to enzymatic digestion in the stomach as well as a poor bioavailability in the small intestine. For these difficult formulations, the encapsulation into nanocarriers would protect the sensitive drug and thus could considerably improve the efficiency of oral drug delivery. In the last years, many candidate biodegradable nanomaterials for such carrier systems have been published. However, before the cargo can be released, the nanocarrier needs to cross multiple barriers of the human body, including a layer of intestinal mucus and epithelial as well as endothelial cells. For overcoming these cellular barriers, transcytosis is favored over a paracellular transport for most nanomaterials as paracellular transport routes lack selectivity of transported molecules once opened up. The exact mechanisms behind the transcellular translocations are up to now still not completely understood. For the vast majority of nanocarriers, the rate of transcellular transport is not sufficient to realize their application in oral drug delivery. Especially trafficking into the endolysosomal pathway often marks a key problem. In this review, we focus on the molecular mechanisms of overcoming cellular barriers, especially transcytosis, and highlight difficulties of oral drug delivery via nanocarriers.
Collapse
Affiliation(s)
- Jonas Reinholz
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Volker Mailänder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| |
Collapse
|
44
|
Suzuki H, Hosomi K, Nasu A, Kondoh M, Kunisawa J. Development of Adjuvant-Free Bivalent Food Poisoning Vaccine by Augmenting the Antigenicity of Clostridium perfringens Enterotoxin. Front Immunol 2018; 9:2320. [PMID: 30356722 PMCID: PMC6189403 DOI: 10.3389/fimmu.2018.02320] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/18/2018] [Indexed: 12/28/2022] Open
Abstract
Clostridium perfringens enterotoxin (CPE) is a common cause of food poisoning and hyperkalemia-associated death. Previously, we reported that fusion of pneumococcal surface protein A (PspA) to C-terminal fragment of CPE (C-CPE) efficiently bound mucosal epithelium so that PspA-specific immune responses could be provoked. In this study, we found that fusion of C-CPE with PspA augmented the antigenicity of C-CPE itself. These findings allowed us to hypothesize that fusion of C-CPE and another food poisoning vaccine act as a bivalent food poisoning vaccine. Therefore, we constructed an adjuvant-free bivalent vaccine against CPE and cholera toxin (CT), which is a major food poisoning in developing country, by genetically fusing CT B subunit to C-CPE. Because of the low antigenicity of C-CPE, immunization of mice with C-CPE alone did not induce C-CPE-specific immune responses. However, immunization with our vaccine induced both C-CPE- and CT-specific neutralizing antibody. The underlying mechanism of the augmented antigenicity of C-CPE included the activation of T cells by CTB. Moreover, neutralizing antibodies lasted for at least 48 weeks and the quality of the antibody was dependent on the binding activity of CTB–C-CPE to its receptors. These findings suggest that our fusion protein is a potential platform for the development of an adjuvant-free bivalent vaccine against CPE and CT.
Collapse
Affiliation(s)
- Hidehiko Suzuki
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Ayaka Nasu
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Sciences, The University of Tokyo, Tokyo, Japan.,Department of Microbiology and Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan.,Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Suita, Japan
| |
Collapse
|
45
|
Gänger S, Schindowski K. Tailoring Formulations for Intranasal Nose-to-Brain Delivery: A Review on Architecture, Physico-Chemical Characteristics and Mucociliary Clearance of the Nasal Olfactory Mucosa. Pharmaceutics 2018; 10:pharmaceutics10030116. [PMID: 30081536 PMCID: PMC6161189 DOI: 10.3390/pharmaceutics10030116] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/25/2018] [Accepted: 08/01/2018] [Indexed: 12/17/2022] Open
Abstract
The blood-brain barrier and the blood-cerebrospinal fluid barrier are major obstacles in central nervous system (CNS) drug delivery, since they block most molecules from entering the brain. Alternative drug delivery routes like intraparenchymal or intrathecal are invasive methods with a remaining risk of infections. In contrast, nose-to-brain delivery is a minimally invasive drug administration pathway, which bypasses the blood-brain barrier as the drug is directed from the nasal cavity to the brain. In particular, the skull base located at the roof of the nasal cavity is in close vicinity to the CNS. This area is covered with olfactory mucosa. To design and tailor suitable formulations for nose-to-brain drug delivery, the architecture, structure and physico-chemical characteristics of the mucosa are important criteria. Hence, here we review the state-of-the-art knowledge about the characteristics of the nasal and, in particular, the olfactory mucosa needed for a rational design of intranasal formulations and dosage forms. Also, the information is suitable for the development of systemic or local intranasal drug delivery as well as for intranasal vaccinations.
Collapse
Affiliation(s)
- Stella Gänger
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Hubertus-Liebrecht-Strasse 35, 88400 Biberach, Germany.
- Faculty of Medicine, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Hubertus-Liebrecht-Strasse 35, 88400 Biberach, Germany.
| |
Collapse
|
46
|
Aithal A, Rauth S, Kshirsagar P, Shah A, Lakshmanan I, Junker WM, Jain M, Ponnusamy MP, Batra SK. MUC16 as a novel target for cancer therapy. Expert Opin Ther Targets 2018; 22:675-686. [PMID: 29999426 PMCID: PMC6300140 DOI: 10.1080/14728222.2018.1498845] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION MUC16 is overexpressed in multiple cancers and plays an important role in tumorigenicity and acquired resistance to therapy. Area covered: In this review, we describe the role of MUC16 under normal physiological conditions and during tumorigenesis. First, we provide a summary of research on MUC16 from its discovery as CA125 to present anti-MUC16 therapy trials that are currently in the initial phases of clinical testing. Finally, we discuss the reasons for the limited effectiveness of these therapies and discuss the direction and focus of future research. Expert opinion: Apart from its protective role in normal physiology, MUC16 contributes to disease progression and metastasis in several malignancies. Due to its aberrant overexpression, it is a promising target for diagnosis and therapy. Cleavage and shedding of its extracellular domain is the major barrier for efficient targeting of MUC16-expressing cancers. Concerted efforts should be undertaken to target the noncleaved cell surface retained portion of MUC16. Such efforts should be accompanied by basic research to understand MUC16 cleavage and decipher the functioning of MUC16 cytoplasmic tail. While previous efforts to activate anti-MUC16 immune response using anti-CA125 idiotype antibodies have met with limited success, ideification of neo-antigenic epitopes in MUC16 that correlate with improved survival have raised raised hopes for developing MUC16-targeted immunotherapy.
Collapse
Affiliation(s)
- Abhijit Aithal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Prakash Kshirsagar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Imayavaramban Lakshmanan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Wade M. Junker
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Moorthy P. Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States of America
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States of America
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States of America
| |
Collapse
|
47
|
Managuli RS, Raut SY, Reddy MS, Mutalik S. Targeting the intestinal lymphatic system: a versatile path for enhanced oral bioavailability of drugs. Expert Opin Drug Deliv 2018; 15:787-804. [PMID: 30025212 DOI: 10.1080/17425247.2018.1503249] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The major challenge of first pass metabolism in oral drug delivery can be surmounted by directing delivery toward intestinal lymphatic system (ILS). ILS circumvents the liver and transports drug directly into systemic circulation via thoracic duct. Lipid and polymeric nanoparticles are transported into ILS through lacteal and Peyer's patches. Moreover, surface modification of nanoparticles with ligand which is specific for Peyer's patches enhances the uptake of drugs into ILS. Bioavailability enhancement by lymphatic uptake is an advantageous approach adopted by scientists today. Therefore, it is important to understand clear insight of ILS in targeted drug delivery and challenges involved in it. AREAS COVERED Current review includes an overview of ILS, factors governing lymphatic transport of nanoparticles and absorption mechanism of lipid and polymeric nanoparticles into ILS. Various ligands used to target Peyer's patch and their conjugation strategies to nanoparticles are explained in detail. In vitro and in vivo models used to assess intestinal lymphatic transport of molecules are discussed further. EXPERT OPINION Although ILS offers a versatile pathway for nanotechnology based targeted drug delivery, extensive investigations on validation of the lymphatic transport models and on the strategies for gastric protection of targeted nanocarriers have to be perceived in for excellent performance of ILS in oral drug delivery.
Collapse
Affiliation(s)
- Renuka Suresh Managuli
- a Department of Pharmaceutics , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal Karnataka State , India
| | - Sushil Yadaorao Raut
- a Department of Pharmaceutics , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal Karnataka State , India
| | - Meka Sreenivasa Reddy
- a Department of Pharmaceutics , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal Karnataka State , India
| | - Srinivas Mutalik
- a Department of Pharmaceutics , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal Karnataka State , India
| |
Collapse
|
48
|
Younes M, Aggett P, Aguilar F, Crebelli R, Dusemund B, Filipič M, Frutos MJ, Galtier P, Gott D, Gundert‐Remy U, Kuhnle GG, Lambré C, Leblanc J, Lillegaard IT, Moldeus P, Mortensen A, Oskarsson A, Stankovic I, Waalkens‐Berendsen I, Wright M, Lodi F, Rincon AM, Smeraldi C, Woutersen RA. Evaluation of four new studies on the potential toxicity of titanium dioxide used as a food additive (E 171). EFSA J 2018; 16:e05366. [PMID: 32625996 PMCID: PMC7009373 DOI: 10.2903/j.efsa.2018.5366] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The European Commission requested EFSA to carry out a scientific evaluation on four studies on the potential toxicity of titanium dioxide (TiO2) used as a food additive (E 171) and to indicate whether they would merit re‐opening the existing opinion of EFSA on the safety of TiO2 (E 171) as a food additive. The results of the Bettini et al. (2017) study did not provide enough justification for a new carcinogenicity study, but, should additional useful mechanistic information become available, this could be reconsidered in future. The new in vitro findings in the Proquin et al. (2017) study did not modify the conclusion on the genotoxicity of TiO2 as stated in the previous EFSA opinion of 2016 on the safety of TiO2 (E 171) as a food additive. The effects of engineered TiO2 nanoparticles reported by the Guo et al. (2017) study were of uncertain biological significance and therefore of limited relevance for the risk assessment of the food additive TiO2 (E 171). There was significant uncertainty in the risk assessment performed by Heringa et al. (2016), which did not include a weight of evidence analysis of the whole database. The Panel considered that the four studies evaluated, highlighted some concerns but with uncertainties, therefore their relevance for the risk assessment was considered limited and further research would be needed to decrease the level of uncertainties. Overall, three of the studies, reporting that TiO2 induced various effects in in vitro and in vivo models, may be useful for hazard identification of TiO2. In the fourth study by Heringa et al. (2016), numerous assumptions were made, which resulted in large uncertainty in their conclusion. Altogether, the Panel concluded that the outcome of the four studies did not merit re‐opening the existing opinion of EFSA related to the safety of TiO2 (E 171) as a food additive.
Collapse
|
49
|
Rios-Arce ND, Collins FL, Schepper JD, Steury MD, Raehtz S, Mallin H, Schoenherr DT, Parameswaran N, McCabe LR. Epithelial Barrier Function in Gut-Bone Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1033:151-183. [PMID: 29101655 DOI: 10.1007/978-3-319-66653-2_8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The intestinal epithelial barrier plays an essential role in maintaining host homeostasis. The barrier regulates nutrient absorption as well as prevents the invasion of pathogenic bacteria in the host. It is composed of epithelial cells, tight junctions, and a mucus layer. Several factors, such as cytokines, diet, and diseases, can affect this barrier. These factors have been shown to increase intestinal permeability, inflammation, and translocation of pathogenic bacteria. In addition, dysregulation of the epithelial barrier can result in inflammatory diseases such as inflammatory bowel disease. Our lab and others have also shown that barrier disruption can have systemic effects including bone loss. In this chapter, we will discuss the current literature to understand the link between intestinal barrier and bone. We will discuss how inflammation, aging, dysbiosis, and metabolic diseases can affect intestinal barrier-bone link. In addition, we will highlight the current suggested mechanism between intestinal barrier and bone.
Collapse
Affiliation(s)
- Naiomy Deliz Rios-Arce
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA.,Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Fraser L Collins
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | | | - Michael D Steury
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Sandi Raehtz
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Heather Mallin
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Danny T Schoenherr
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Narayanan Parameswaran
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA. .,Department of Physiology, Michigan State University, East Lansing, MI, USA.
| | - Laura R McCabe
- Department of Physiology and Department of Radiology, Biomedical Imaging Research Centre, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
50
|
Diesner SC, Bergmayr C, Wang XY, Heiden D, Exenberger S, Roth-Walter F, Starkl P, Ret D, Pali-Schöll I, Gabor F, Untersmayr E. Characterization of Vibrio cholerae neuraminidase as an immunomodulator for novel formulation of oral allergy immunotherapy. Clin Immunol 2018; 192:30-39. [PMID: 29608970 DOI: 10.1016/j.clim.2018.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 09/05/2017] [Accepted: 03/28/2018] [Indexed: 01/22/2023]
Abstract
To improve current mucosal allergen immunotherapy Vibrio cholerae neuraminidase (NA) was evaluated as a novel epithelial targeting molecule for functionalization of allergen-loaded, poly(D,L-lactide-co-glycolide) (PLGA) microparticles (MPs) and compared to the previously described epithelial targeting lectins wheat germ agglutinin (WGA) and Aleuria aurantia lectin (AAL). All targeters revealed binding to Caco-2 cells, but only NA had high binding specificity to α-L fucose and monosialoganglioside-1. An increased transepithelial uptake was found for NA-MPs in a M-cell co-culture model. NA and NA-MPs induced high levels of IFN-γ and IL10 in naive mouse splenocytes and CCL20 expression in Caco-2. Repeated oral gavage of NA-MPs resulted in a modulated, allergen-specific immune response. In conclusion, NA has enhanced M-cell specificity compared to the other targeters. NA functionalized MPs induce a Th1 and T-regulatory driven immune response and avoid allergy effector cell activation. Therefore, it is a promising novel, orally applied formula for allergy therapy.
Collapse
Affiliation(s)
- Susanne C Diesner
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Cornelia Bergmayr
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Xue-Yan Wang
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Denise Heiden
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Sarah Exenberger
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Franziska Roth-Walter
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Philipp Starkl
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Davide Ret
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria
| | - Isabella Pali-Schöll
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Franz Gabor
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|