1
|
Lu J, Liu X, Li W, Xi C, Feng D, Song S. Analysis of the sensitization activity of Moringa oleifera leaves protein. Front Nutr 2025; 11:1509343. [PMID: 39912058 PMCID: PMC11794069 DOI: 10.3389/fnut.2024.1509343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/18/2024] [Indexed: 02/07/2025] Open
Abstract
The determination of allergenic proteins in Moringa oleifera leaves, which is the main components of immune activity, has enabled the development of a more effective method for evaluating the activity of extracted Moringa oleifera leaves protein. In this study, the extraction process of Moringa oleifera leaves protein was optimized based on a single factor experiment. The hemagglutination-related properties of Moringa oleifera leaves protein, such as (thermal, acid-base) stability, sugar binding specificity, ion binding characteristics, and hemolytic activity, were detected. The optimal combination of extraction process was: extraction time of 6 h, material-liquid ratio of 1:8, and ammonium sulfate saturation of 60%. The extraction rate of moringa leaf protein under this condition was 14.37 mg/g. The molecular weight of moringa leaf protein was analyzed by SDS-PAGE, and the molecular weight was mainly concentrated around 23 kDa~70 kDa, with the highest content of 35 kDa (major allergen). The study of the hemagglutination characteristics of Moringa oleifera leaves protein revealed that the protein exhibited high stability at temperatures below 60°C, with complete loss of activity occurring at temperatures above 110°C for 20 min. The effect of different pH conditions on the hemagglutination capacity of Moringa oleifera leaves protein was readily discernible. The hemagglutination activity of Moringa oleifera leaves protein was 104 in a pH value from 3.7 to 7.8, and the hemagglutination activity was completely lost at a pH value higher than 11.9. D(+) anhydrous glucose is the specific inhibitory sugar of Moringa oleifera leaves protein lectin. Moringa oleifera leaves protein exhibits hemolytic activity at a concentration of at least 20 mg/mL, and α-methyl-mannoside, galactoside, raffinose and Al3+ can inhibit the hemolysis of Moringa oleifera leaves protein. The present study identified the effects of different factors on the coagulation activity and hemolytic ability of Moringa oleifera leaves protein, thereby providing a theoretical basis for further purification and application of Moringa oleifera lectin. However, it should be noted that the results of the mixture have certain limitations, and further purification of lectin is needed to obtain more targeted research results.
Collapse
Affiliation(s)
- Juan Lu
- Yunnan Agricultural University, Kunming, China
- Yunnan Provincial High Court Characteristic Agricultural Industry Research Institute, Kunming, China
| | - Xiaoxue Liu
- Yunnan Agricultural University, Kunming, China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Wenjie Li
- Yunnan Agricultural University, Kunming, China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Chuyu Xi
- Yunnan Agricultural University, Kunming, China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Dan Feng
- Yunnan Agricultural University, Kunming, China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Shuang Song
- Yunnan Agricultural University, Kunming, China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
2
|
Silva MLS. Lectin-modified drug delivery systems - Recent applications in the oncology field. Int J Pharm 2024; 665:124685. [PMID: 39260750 DOI: 10.1016/j.ijpharm.2024.124685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Chemotherapy with cytotoxic drugs remains the core treatment for cancer but, due to the difficulty to find general and usable biochemical differences between cancer cells and normal cells, many of these drugs are associated with lack of specificity, resulting in side effects and collateral cytotoxicity that impair patients' adherence to therapy. Novel cancer treatments in which the cytotoxic effect is maximized while adverse effects are reduced can be implemented by developing targeted therapies that exploit the specific features of cancer cells, such as the typical expression of aberrant glycans. Modification of drug delivery systems with lectins is one of the strategies to implement targeted chemotherapies, as lectins are able to specifically recognize and bind to cancer-associated glycans expressed at the surface of cancer cells, guiding the drug treatment towards these cells and not affecting healthy ones. In this paper, recent advances on the development of lectin-modified drug delivery systems for targeted cancer treatments are thoroughly reviewed, with a focus on their properties and performance in diverse applications, as well as their main advantages and limitations. The synthesis and analytical characterization of the cited lectin-modified drug delivery systems is also briefly described. A comparison with free-drug treatments and with antibody-modified drug delivery systems is presented, emphasizing the advantages of lectin-modified drug delivery systems. Main constraints and potential challenges of lectin-modified drug delivery systems, including key difficulties for clinical translation of these systems, and the required developments in this area, are also signalled.
Collapse
Affiliation(s)
- Maria Luísa S Silva
- Centro de Estudos Globais, Universidade Aberta, Rua da Escola Politécnica 147, 1269-001 Lisboa, Portugal.
| |
Collapse
|
3
|
Kumar A, Sharma A, Tirpude NV, Padwad Y, Hallan V, Kumar S. Plant-derived immuno-adjuvants in vaccines formulation: a promising avenue for improving vaccines efficacy against SARS-CoV-2 virus. Pharmacol Rep 2022; 74:1238-1254. [PMID: 36125739 PMCID: PMC9487851 DOI: 10.1007/s43440-022-00418-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 12/13/2022]
Abstract
The SARS-CoV-2 outbreak has posed a plethora of problems for the global healthcare system and socioeconomic burden. Despite valiant efforts to contain the COVID-19 outbreak, the situation has deteriorated to the point that there are no viable preventive therapies to treat this disease. The case count has skyrocketed globally due to the newly evolved variants. Despite vaccination drives, the re-occurrence of recent pandemic waves has reinforced the importance of innovation/utilization of immune-booster to achieve appropriate long-term vaccine protection. Plant-derived immuno-adjuvants, which have multifaceted functions, can impede infections by boosting the immune system. Many previous studies have shown that formulation of vaccines using plant-derived adjuvant results in long-lasting immunity may overcome the natural tendency of coronavirus immunity to wane quickly. Plant polysaccharides, glycosides, and glycoprotein extracts have reportedly been utilized as enticing adjuvants in experimental vaccines, such as Advax, Matrix-M, and Mistletoe lectin, which have been shown to be highly immunogenic and safe. When employed in vaccine formulation, Advax and Matrix-M generate long-lasting antibodies, a balanced robust Th1/Th2 cytokine profile, and the stimulation of cytotoxic T cells. Thus, the use of adjuvants derived from plants may increase the effectiveness of vaccines, resulting in the proper immunological response required to combat COVID-19. A few have been widely used in epidemic outbreaks, including SARS and H1N1 influenza, and their use could also improve the efficacy of COVID-19 vaccines. In this review, the immunological adjuvant properties of plant compounds as well as their potential application in anti-COVID-19 therapy are thoroughly discussed.
Collapse
Affiliation(s)
- Arbind Kumar
- COVID-19 Testing facility, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Aashish Sharma
- COVID-19 Testing facility, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Narendra Vijay Tirpude
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Yogendra Padwad
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Vipin Hallan
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Sanjay Kumar
- CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| |
Collapse
|
4
|
Nazeam JA, Singab ANB. Immunostimulant plant proteins: Potential candidates as vaccine adjuvants. Phytother Res 2022; 36:4345-4360. [PMID: 36128599 PMCID: PMC9538006 DOI: 10.1002/ptr.7624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 08/20/2022] [Accepted: 09/03/2022] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic is shaking up global scientific structures toward addressing antibiotic resistance threats and indicates an urgent need to develop more cost-effective vaccines. Vaccine adjuvants play a crucial role in boosting immunogenicity and improving vaccine efficacy. The toxicity and adversity of most adjuvant formulations are the major human immunization problems, especially in routine pediatric and immunocompromised patients. The present review focused on preclinical studies of immunoadjuvant plant proteins in use with antiparasitic, antifungal, and antiviral vaccines. Moreover, this report outlines the current perspective of immunostimulant plant protein candidates that can be used by researchers in developing new generations of vaccine-adjuvants. Future clinical studies are required to substantiate the plant proteins' safety and applicability as a vaccine adjuvant in pharmaceutical manufacturing.
Collapse
Affiliation(s)
- Jilan A. Nazeam
- Pharmacognosy Department, Faculty of PharmacyOctober 6 UniversityGizaEgypt
| | | |
Collapse
|
5
|
Padiyappa SD, Avalappa H, Somegowda M, Sridhara S, Venkatesh YP, Prabhakar BT, Pramod SN, Almujaydil MS, Shokralla S, Abdelbacki AMM, Elansary HO, El-Sabrout AM, Mahmoud EA. Immunoadjuvant and Humoral Immune Responses of Garlic ( Allium sativum L.) Lectins upon Systemic and Mucosal Administration in BALB/c Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041375. [PMID: 35209158 PMCID: PMC8880535 DOI: 10.3390/molecules27041375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/31/2022] [Accepted: 02/11/2022] [Indexed: 12/12/2022]
Abstract
Dietary food components have the ability to affect immune function; following absorption, specifically orally ingested dietary food containing lectins can systemically modulate the immune cells and affect the response to self- and co-administered food antigens. The mannose-binding lectins from garlic (Allium sativum agglutinins; ASAs) were identified as immunodulatory proteins in vitro. The objective of the present study was to assess the immunogenicity and adjuvanticity of garlic agglutinins and to evaluate whether they have adjuvant properties in vivo for a weak antigen ovalbumin (OVA). Garlic lectins (ASA I and ASA II) were administered by intranasal (50 days duration) and intradermal (14 days duration) routes, and the anti-lectin and anti-OVA immune (IgG) responses in the control and test groups of the BALB/c mice were assessed for humoral immunogenicity. Lectins, co-administered with OVA, were examined for lectin-induced anti-OVA IgG response to assess their adjuvant properties. The splenic and thymic indices were evaluated as a measure of immunomodulatory functions. Intradermal administration of ASA I and ASA II had showed a four-fold and two-fold increase in anti-lectin IgG response, respectively, vs. the control on day 14. In the intranasal route, the increases were 3-fold and 2.4-fold for ASA I and ASA II, respectively, on day 50. No decrease in the body weights of animals was noticed; the increases in the spleen and thymus weights, as well as their indices, were significant in the lectin groups. In the adjuvanticity study by intranasal administration, ASA I co-administered with ovalbumin (OVA) induced a remarkable increase in anti-OVA IgG response (~six-fold; p < 0.001) compared to the control, and ASA II induced a four-fold increase vs. the control on day 50. The results indicated that ASA was a potent immunogen which induced mucosal immunogenicity to the antigens that were administered intranasally in BALB/c mice. The observations made of the in vivo study indicate that ASA I has the potential use as an oral and mucosal adjuvant to deliver candidate weak antigens. Further clinical studies in humans are required to confirm its applicability.
Collapse
Affiliation(s)
- Shruthishree D. Padiyappa
- Food Allergy and Immunology Laboratory, Department of Studies in Food Technology, Davangere University, Shivagangotri, Davangere 577 007, India; (S.D.P.); (H.A.)
- Molecular Biomedicine Laboratory, Postgraduate Department of Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga 577 203, India;
| | - Hemavathi Avalappa
- Food Allergy and Immunology Laboratory, Department of Studies in Food Technology, Davangere University, Shivagangotri, Davangere 577 007, India; (S.D.P.); (H.A.)
- Molecular Biomedicine Laboratory, Postgraduate Department of Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga 577 203, India;
| | - Madhusudana Somegowda
- Department of Plant Biochemistry, University of Agriculture and Horticulture Science, Shivamogga 577 204, India;
| | - Shankarappa Sridhara
- Center for Climate Resilient Agriculture, University of Agriculture and Horticulture Science, Shivamogga 577 204, India;
| | - Yeldur P. Venkatesh
- Department of Biochemistry and Nutrition, CSIR–Central Food Technological Research Institute (CFTRI), Mysuru 570 020, India;
| | - Bettadatunga T. Prabhakar
- Molecular Biomedicine Laboratory, Postgraduate Department of Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga 577 203, India;
| | - Siddanakoppalu N. Pramod
- Food Allergy and Immunology Laboratory, Department of Studies in Food Technology, Davangere University, Shivagangotri, Davangere 577 007, India; (S.D.P.); (H.A.)
- Correspondence: (S.N.P.); (H.O.E.)
| | - Mona S. Almujaydil
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Shadi Shokralla
- Centre for Biodiversity Genomics, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Ashraf M. M. Abdelbacki
- Applied Studies and Community Service College, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Hosam O. Elansary
- Plant Production Department, College of Food & Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
- Correspondence: (S.N.P.); (H.O.E.)
| | - Ahmed M. El-Sabrout
- Department of Applied Entomology and Zoology, Faculty of Agriculture (EL-Shatby), Alexandria University, Alexandria 21545, Egypt;
| | - Eman A. Mahmoud
- Department of Food Industries, Faculty of Agriculture, Damietta University, Damietta 34511, Egypt;
| |
Collapse
|
6
|
Wang Y, He S, Zhou F, Sun H, Cao X, Ye Y, Li J. Detection of Lectin Protein Allergen of Kidney Beans ( Phaseolus vulgaris L.) and Desensitization Food Processing Technology. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14723-14741. [PMID: 34251800 DOI: 10.1021/acs.jafc.1c02801] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
With the increase of food allergy events related to not properly cooked kidney beans (Phaseolus vulgaris L.), more and more researchers are paying attention to the sensitization potential of lectin, one of the major storage and defensive proteins with the specific carbohydrate-binding activity. The immunoglobulin E (IgE), non-IgE, and mixed allergic reactions induced by the lectins were inducted in the current paper, and the detection methods of kidney bean lectin, including the purification strategies, hemagglutination activity, specific polysaccharide or glycoprotein interactions, antibody combinations, mass spectrometry methods, and allergomics strategies, were summarized, while various food processing aspects, such as the physical thermal processing, physical non-thermal processing, chemical modifications, and biological treatments, were reviewed in the potential of sensitization reduction. It might be the first comprehensive review on lectin allergen detection from kidney bean and the desensitization strategy in food processing and will provide a basis for food safety control.
Collapse
Affiliation(s)
- Yongfei Wang
- Engineering Research Center of Bio-process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Shudong He
- Engineering Research Center of Bio-process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Fanlin Zhou
- Engineering Research Center of Bio-process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Hanju Sun
- Engineering Research Center of Bio-process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Xiaodong Cao
- Engineering Research Center of Bio-process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Yongkang Ye
- Engineering Research Center of Bio-process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Jing Li
- College of Biological and Environmental Engineering, Hefei University, Hefei, Anhui 230601, People's Republic of China
| |
Collapse
|
7
|
Butle AB, Talmale SA, Jadhao VV, Patil MB, Lambat TL. Immunostimulatory and anti-allergic potential of novel heterotrimeric lectin from seeds of Zizyphus mauritiana Lam. Int J Biol Macromol 2021; 171:389-397. [PMID: 33428960 DOI: 10.1016/j.ijbiomac.2021.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
Zizyphus mauritiana Lam. seeds (ZMS) have been used medicinally as sedative or hypnotic drugs in most of Asian countries. ZMS has significant benefits to the human health. Therefore, we have evaluated immunomodulatory effect of lectin extracted from these ZMSL in both in vitro and in vivo study. Anaphylaxis is a severe life-threatening allergic reaction and Arthus reaction is deposition of immune complex and complement system activation, so we hypothesized that if ZMSL can protect these severe allergic diseases. We have studied the effect of ZMSL on macrophages and Wistar albino rats and confirmed its protective effect against anaphylaxis and Arthus reaction. Results of this study suggest ZMSL have immunostimulatory and antiallergic activity.
Collapse
Affiliation(s)
- Ashwin B Butle
- Department of Biochemistry, RashtrasantTukadoji Maharaj Nagpur University, Nagpur 440033, Maharashtra, India.
| | - Suhas A Talmale
- Department of Biochemistry, RashtrasantTukadoji Maharaj Nagpur University, Nagpur 440033, Maharashtra, India
| | - Vivek V Jadhao
- Department of Biochemistry, RashtrasantTukadoji Maharaj Nagpur University, Nagpur 440033, Maharashtra, India
| | - Mandakini B Patil
- Department of Biochemistry, RashtrasantTukadoji Maharaj Nagpur University, Nagpur 440033, Maharashtra, India.
| | - Trimurti L Lambat
- Department of Chemistry, Manoharbhai Patel College of Arts, Commerce & Science, Deori, dist-Gondia 441901, Maharashtra, India.
| |
Collapse
|
8
|
Farr AC, Xiong MP. Challenges and Opportunities of Deferoxamine Delivery for Treatment of Alzheimer's Disease, Parkinson's Disease, and Intracerebral Hemorrhage. Mol Pharm 2020; 18:593-609. [PMID: 32926630 DOI: 10.1021/acs.molpharmaceut.0c00474] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Deferoxamine mesylate (DFO) is an FDA-approved, hexadentate iron chelator routinely used to alleviate systemic iron burden in thalassemia major and sickle cell patients. Iron accumulation in these disease states results from the repeated blood transfusions required to manage these conditions. Iron accumulation has also been implicated in the pathogenesis of Alzheimer's disease (AD), Parkinson's disease (PD), and secondary injury following intracerebral hemorrhage (ICH). Chelation of brain iron is thus a promising therapeutic strategy for improving behavioral outcomes and slowing neurodegeneration in the aforementioned disease states, though the effectiveness of DFO treatment is limited on several accounts. Systemically administered DFO results in nonspecific toxicity at high doses, and the drug's short half-life leads to low patient compliance. Mixed reports of DFO's ability to cross the blood-brain barrier (BBB) also appear in literature. These limitations necessitate novel DFO formulations prior to the drug's widespread use in managing neurodegeneration. Herein, we discuss the various dosing regimens and formulations employed in intranasal (IN) or systemic DFO treatment, as well as the physiological and behavioral outcomes observed in animal models of AD, PD, and ICH. The clinical progress of chelation therapy with DFO in managing neurodegeneration is also evaluated. Finally, the elimination of intranasally administered particles via the glymphatic system and efflux transporters is discussed. Abundant preclinical evidence suggests that intranasal DFO treatment improves memory retention and behavioral outcome in rodent models of AD, PD, and ICH. Several other biochemical and physiological metrics, such as tau phosphorylation, the survival of tyrosine hydroxylase-positive neurons, and infarct volume, are also positively affected by intranasal DFO treatment. However, dosing regimens are inconsistent across studies, and little is known about brain DFO concentration following treatment. Systemic DFO treatment yields similar results, and some complex formulations have been developed to improve permeability across the BBB. However, despite the success in preclinical models, clinical translation is limited with most clinical evidence investigating DFO treatment in ICH patients, where high-dose treatment has proven dangerous and dosing regimens are not consistent across studies. DFO is a strong drug candidate for managing neurodegeneration in the aging population, but before it can be routinely implemented as a therapeutic agent, dosing regimens must be standardized, and brain DFO content following drug administration must be understood and controlled via novel formulations.
Collapse
Affiliation(s)
- Amy Corbin Farr
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - May P Xiong
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
9
|
Giglio ML, Ituarte S, Ibañez AE, Dreon MS, Prieto E, Fernández PE, Heras H. Novel Role for Animal Innate Immune Molecules: Enterotoxic Activity of a Snail Egg MACPF-Toxin. Front Immunol 2020; 11:428. [PMID: 32231667 PMCID: PMC7082926 DOI: 10.3389/fimmu.2020.00428] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/25/2020] [Indexed: 01/22/2023] Open
Abstract
Gastropod Molluscs rely exclusively on the innate immune system to protect from pathogens, defending their embryos through maternally transferred effectors. In this regard, Pomacea snail eggs, in addition to immune defenses, have evolved the perivitellin-2 or PV2 combining two immune proteins into a neurotoxin: a lectin and a pore-forming protein from the Membrane Attack Complex/Perforin (MACPF) family. This binary structure resembles AB-toxins, a group of toxins otherwise restricted to bacteria and plants. Many of these are enterotoxins, leading us to explore this activity in PV2. Enterotoxins found in bacteria and plants act mainly as pore-forming toxins and toxic lectins, respectively. In animals, although both pore-forming proteins and lectins are ubiquitous, no enterotoxins have been reported. Considering that Pomacea snail eggs ingestion induce morpho-physiological changes in the intestinal mucosa of rodents and is cytotoxic to intestinal cells in culture, we seek for the factor causing these effects and identified PmPV2 from Pomacea maculata eggs. We characterized the enterotoxic activity of PmPV2 through in vitro and in vivo assays. We determined that it withstands the gastrointestinal environment and resisted a wide pH range and enzymatic proteolysis. After binding to Caco-2 cells it promoted changes in surface morphology and an increase in membrane roughness. It was also cytotoxic to both epithelial and immune cells from the digestive system of mammals. It induced enterocyte death by a lytic mechanism and disrupted enterocyte monolayers in a dose-dependent manner. Further, after oral administration to mice PmPV2 attached to enterocytes and induced large dose-dependent morphological changes on their small intestine mucosa, reducing the absorptive surface. Additionally, PmPV2 was detected in the Peyer's patches where it activated lymphoid follicles and triggered apoptosis. We also provide evidence that the toxin can traverse the intestinal barrier and induce oral adaptive immunity with evidence of circulating antibody response. As a whole, these results indicate that PmPV2 is a true enterotoxin, a role that has never been reported to lectins or perforin in animals. This extends by convergent evolution the presence of plant- and bacteria-like enterotoxins to animals, thus expanding the diversity of functions of MACPF proteins in nature.
Collapse
Affiliation(s)
- Matías L Giglio
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner" (INIBIOLP), CONICET, CCT-La Plata, Universidad Nacional de la Plata (UNLP), La Plata, Argentina
| | - Santiago Ituarte
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner" (INIBIOLP), CONICET, CCT-La Plata, Universidad Nacional de la Plata (UNLP), La Plata, Argentina
| | - Andrés E Ibañez
- División de Vertebrados, Facultad de Ciencias Naturales y Museo (FCNyM), Universidad Nacional de La Plata, La Plata, Argentina
| | - Marcos S Dreon
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner" (INIBIOLP), CONICET, CCT-La Plata, Universidad Nacional de la Plata (UNLP), La Plata, Argentina
- Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, Universidad Nacional de la Plata (UNLP), La Plata, Argentina
| | - Eduardo Prieto
- Instituto de Investigaciones Físico-químicas Teóricas y Aplicadas (INIFTA), CONICET, CCT-La Plata, Universidad Nacional de La Plata, La Plata, Argentina
| | - Patricia E Fernández
- Facultad de Ciencias Veterinarias (FEV), Instituto de Patología B. Epstein, Cátedra de Patología General Veterinaria, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Horacio Heras
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner" (INIBIOLP), CONICET, CCT-La Plata, Universidad Nacional de la Plata (UNLP), La Plata, Argentina
| |
Collapse
|
10
|
Wang W, Gong C, Han Z, Lv X, Liu S, Wang L, Song L. The lectin domain containing proteins with mucosal immunity and digestive functions in oyster Crassostrea gigas. FISH & SHELLFISH IMMUNOLOGY 2019; 89:237-247. [PMID: 30936048 DOI: 10.1016/j.fsi.2019.03.067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 06/09/2023]
Abstract
Lectins are carbohydrate-binding proteins with lectin domains, which are extensively studied for their numerous roles in biological recognition. However, the lectin domain containing proteins (LDCPs) chimerized with other non-lectin domains have not received sufficient attention. In the present study, a genome-wide survey of LDCPs in oyster Crassostrea gigas was conducted, and an expansive 640 LDCPs derived from ten lectin domains were identified and functionally explored. In these LDCPs, a total of 282 kinds of domains were predicted, and 90% of the LDCPs contained more than one kind of domain. The lectin domains were frequently fused with non-lectin domains, such as epidermal growth factor domain and peptidase related domains, which supplied LDCPs with more diversity in structures and functions. The C-type lectin domains were the most abundant domains in LDCPs, and they were largely co-existed with non-lectin domains of complement activation-related domains (such as CUB domain and PAN-1 domain) but relative independence with other lectin domains. Furthermore, the C-type lectin domain containing proteins (CTLPs) found to mainly act as pattern immune recognition receptors and were highly expressed in mucosal tissues (digestive gland, male gonad and labial palp) to provide mucosal immune protections. The Concanavalin A-like lectin domains were the second richest domains in LDCPs, and they were mostly constructed into chimeric proteins with epidermal growth factor domain and peptidase related domains. The Concanavalin A-like lectin domain containing proteins (CALPs) were significantly enriched with peptidase activities and mainly expressed in digestive tissues. All the results suggested the mucosal immunity and digestive functions of oyster LDCPs, which provided a fresh idea about the functions of invertebrate lectin family.
Collapse
Affiliation(s)
- Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Changhao Gong
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Zirong Han
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Xiaojing Lv
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Shujing Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
11
|
Sander VA, Corigliano MG, Clemente M. Promising Plant-Derived Adjuvants in the Development of Coccidial Vaccines. Front Vet Sci 2019; 6:20. [PMID: 30809529 PMCID: PMC6379251 DOI: 10.3389/fvets.2019.00020] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/18/2019] [Indexed: 01/15/2023] Open
Abstract
Coccidial parasites cause medical and veterinary diseases worldwide, frequently leading to severe illness and important economic losses. At present, drugs, chemotherapeutics and prophylactic vaccines are still missing for most of the coccidial infections. Moreover, the development and administration of drugs and chemotherapeutics against these diseases would not be adequate in livestock, since they may generate unacceptable residues in milk and meat that would avoid their commercialization. In this scenario, prophylactic vaccines emerge as the most suitable approach. Subunit vaccines have proven to be biologically safe and economically viable, allowing researchers to choose among the best antigens against each pathogen. However, they are generally poorly immunogenic and require the addition of adjuvant compounds to the vaccine formulation. During the last decades, research involving plant immunomodulatory compounds has become an important field of study based on their potential pharmaceutical applications. Some plant molecules such as saponins, polysaccharides, lectins and heat shock proteins are being explored as candidates for adjuvant/carriers formulations. Moreover, plant-derived immune stimulatory compounds open the possibility to attain the main goal in adjuvant research: a safe and non-toxic adjuvant capable of strongly boosting and directing immune responses that could be incorporated into different vaccine formulations, including mucosal vaccines. Here, we review the immunomodulatory properties of several plant molecules and discuss their application and future perspective as adjuvants in the development of vaccines against coccidial infections.
Collapse
Affiliation(s)
- Valeria A Sander
- Unidad de Biotecnología 6-UB6, Instituto Tecnológico Chascomús (INTECh), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional de General San Martín (UNSAM), Chascomús, Argentina
| | - Mariana G Corigliano
- Unidad de Biotecnología 6-UB6, Instituto Tecnológico Chascomús (INTECh), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional de General San Martín (UNSAM), Chascomús, Argentina
| | - Marina Clemente
- Unidad de Biotecnología 6-UB6, Instituto Tecnológico Chascomús (INTECh), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional de General San Martín (UNSAM), Chascomús, Argentina
| |
Collapse
|
12
|
He S, Simpson BK, Sun H, Ngadi MO, Ma Y, Huang T. Phaseolus vulgaris lectins: A systematic review of characteristics and health implications. Crit Rev Food Sci Nutr 2017; 58:70-83. [PMID: 26479307 DOI: 10.1080/10408398.2015.1096234] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Legume lectins are carbohydrate-binding proteins of non-immune origin. Significant amounts of lectins have been found in Phaseolus vulgaris beans as far back as in the last century; however, many questions about their potential biological roles still remain obscure. Studies have shown that lectins are anti-nutritional factors that can cause intestinal disorders. Owing to their ability to act as toxic allergens and hemagglutinins, the Phaseolus vulgaris lectins are of grave concern for human health and safety. Nonetheless, their potential beneficial health effects, such as anti-cancer, anti-human immunodeficiency virus (anti-HIV), anti-microbial infection, preventing mucosal atrophy, reducing type 2 diabetes and obesity, promoting nutrients absorption and targeting drugs, are of immense interest. The significance of Phaseolus vulgaris lectins in biological researches and the potential biomedical applications have placed tremendous emphasis on the development of purification strategies to obtain the protein in pure and stable forms. These purification strategies entail considerations such as effects of proteolysis, heating, gamma radiation, and high-hydrostatic-pressure that can have crucial outcomes in either eliminating or improving bioactivities of the lectins. Thus, up-to-date research findings of Phaseolus vulgaris lectins on different aspects such as anti-nutritional and health impacts, purification strategies and novel processing trends, are systematically reviewed.
Collapse
Affiliation(s)
- Shudong He
- a School of Biotechnology and Food Engineering , Hefei University of Technology , Hefei , Anhui , China.,b School of Food Science and Engineering , Harbin Institute of Technology , Harbin , Heilongjiang , China.,c Department of Food Science and Agricultural Chemistry , Macdonald Campus, McGill University , Ste-Anne-de-Bellevue, Québec , Canada
| | - Benjamin K Simpson
- c Department of Food Science and Agricultural Chemistry , Macdonald Campus, McGill University , Ste-Anne-de-Bellevue, Québec , Canada
| | - Hanju Sun
- a School of Biotechnology and Food Engineering , Hefei University of Technology , Hefei , Anhui , China
| | - Michael O Ngadi
- d Department of Bioresource Engineering , Macdonald Campus, McGill University , Ste-Anne-de-Bellevue, Québec , Canada
| | - Ying Ma
- b School of Food Science and Engineering , Harbin Institute of Technology , Harbin , Heilongjiang , China
| | - Tiemin Huang
- e Advanced Electrophoresis Solutions Ltd. , Cambridge , Ontario , Canada
| |
Collapse
|
13
|
Nayak TR, Wang H, Pant A, Zheng M, Junginger H, Goh WJ, Lee CK, Zou S, Alonso S, Czarny B, Storm G, Sow CH, Lee C, Pastorin G. ZnO Nano-Rod Devices for Intradermal Delivery and Immunization. NANOMATERIALS 2017; 7:nano7060147. [PMID: 28617335 PMCID: PMC5485794 DOI: 10.3390/nano7060147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/09/2017] [Accepted: 06/09/2017] [Indexed: 11/23/2022]
Abstract
Intradermal delivery of antigens for vaccination is a very attractive approach since the skin provides a rich network of antigen presenting cells, which aid in stimulating an immune response. Numerous intradermal techniques have been developed to enhance penetration across the skin. However, these methods are invasive and/or affect the skin integrity. Hence, our group has devised zinc oxide (ZnO) nano-rods for non-destructive drug delivery. Chemical vapour deposition was used to fabricate aligned nano-rods on ZnO pre-coated silicon chips. The nano-rods’ length and diameter were found to depend on the temperature, time, quality of sputtered silicon chips, etc. Vertically aligned ZnO nano-rods with lengths of 30–35 µm and diameters of 200–300 nm were selected for in vitro human skin permeation studies using Franz cells with Albumin-fluorescein isothiocyanate (FITC) absorbed on the nano-rods. Fluorescence and confocal studies on the skin samples showed FITC penetration through the skin along the channels formed by the nano-rods. Bradford protein assay on the collected fluid samples indicated a significant quantity of Albumin-FITC in the first 12 h. Low antibody titres were observed with immunisation on Balb/c mice with ovalbumin (OVA) antigen coated on the nano-rod chips. Nonetheless, due to the reduced dimensions of the nano-rods, our device offers the additional advantage of excluding the simultaneous entrance of microbial pathogens. Taken together, these results showed that ZnO nano-rods hold the potential for a safe, non-invasive, and painless intradermal drug delivery.
Collapse
Affiliation(s)
- Tapas R Nayak
- Department of Pharmacy, National University of Singapore, Singapore 117583, Singapore.
| | - Hao Wang
- Department of Electrical Engineering, National University of Singapore, Singapore 117583, Singapore.
| | - Aakansha Pant
- Department of Pharmacy, National University of Singapore, Singapore 117583, Singapore.
| | - Minrui Zheng
- Department of Physics, National University of Singapore, Singapore 117551, Singapore.
| | - Hans Junginger
- Department of Pharmacy, National University of Singapore, Singapore 117583, Singapore.
| | - Wei Jiang Goh
- Department of Pharmacy, National University of Singapore, Singapore 117583, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences (CeLS), Singapore 117456, Singapore.
| | - Choon Keong Lee
- Department of Pharmacy, National University of Singapore, Singapore 117583, Singapore.
| | - Shui Zou
- Department of Pharmacy, National University of Singapore, Singapore 117583, Singapore.
| | - Sylvie Alonso
- Department of Microbiology, National University of Singapore, Singapore 117545, Singapore.
| | - Bertrand Czarny
- School of Materials Science and Engineering (MSE) & Lee Kong Chian School of medicine (LKCmedicine), Nanyang Technological University, Singapore 636921, Singapore.
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| | - Chorng Haur Sow
- Department of Physics, National University of Singapore, Singapore 117551, Singapore.
| | - Chengkuo Lee
- Department of Electrical Engineering, National University of Singapore, Singapore 117583, Singapore.
| | - Giorgia Pastorin
- Department of Pharmacy, National University of Singapore, Singapore 117583, Singapore.
- Department of Physics, National University of Singapore, Singapore 117551, Singapore.
- NUSNNI-NanoCore, National University of Singapore, T-Lab, Blk E3-05-29, 2 Engineering Drive 3, Singapore 117581, Singapore.
| |
Collapse
|
14
|
Chitosan-catechol: A polymer with long-lasting mucoadhesive properties. Biomaterials 2015; 52:161-70. [DOI: 10.1016/j.biomaterials.2015.02.010] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 01/30/2015] [Accepted: 02/01/2015] [Indexed: 01/03/2023]
|
15
|
Hou J, Liu Y, Hsi J, Wang H, Tao R, Shao Y. Cholera toxin B subunit acts as a potent systemic adjuvant for HIV-1 DNA vaccination intramuscularly in mice. Hum Vaccin Immunother 2014; 10:1274-83. [PMID: 24633335 PMCID: PMC4896579 DOI: 10.4161/hv.28371] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 02/17/2014] [Accepted: 02/27/2014] [Indexed: 11/19/2022] Open
Abstract
Cholera toxin B subunit (CTB) was investigated as a classical mucosal adjuvant that can increase vaccine immunogenicity. In this study, we found out the in vitro efficacy of cholera toxin B subunit (CTB) in activating mice bone marrow-derived dendritic cells (BMDCs) through Toll-like receptor signaling pathways. In vitro RNA and transcriptional level profiling arrays revealed that CTB guides high levels of Th1 and Th2 type cytokines, inflammatory cytokines, and chemokines. Based on the robustness of these profiling results, we examined the induction of HIV Env-specific immunity by CTB co-inoculated with HIV Env DNA vaccine intramuscularly in vivo. CTB enhanced HIV-Env specific cellular immune responses in Env-specific IFN-γ ELISPOT, compared with DNA vaccine alone. Moreover, CTB induced high levels of Env specific humoral response and promoted antibody maturation after the third round of vaccination. This combination immunization strategy induced a Th2-type bias response which is indicative of a high ratio of IgG1/IgG2a. This study reports that CTB as a classical mucosal adjuvant could enhance HIV-1 DNA-based vaccine immunogenicity intramuscularly; therefore, these findings suggest that CTB could serve as an effective candidate adjuvant for DNA vaccination.
Collapse
Affiliation(s)
- Jue Hou
- State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention; Beijing, PR China
- Center of Medical Physics and Technology; Hefei Institutes of Physical Science; Chinese Academy of Sciences; Hefei, PR China
| | - Ying Liu
- State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention; Beijing, PR China
| | - Jenny Hsi
- State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention; Beijing, PR China
| | - Hongzhi Wang
- Center of Medical Physics and Technology; Hefei Institutes of Physical Science; Chinese Academy of Sciences; Hefei, PR China
| | - Ran Tao
- The Fourth Affiliated Hospital of Anhui Medical University; Hefei, PR China
| | - Yiming Shao
- State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention; Beijing, PR China
| |
Collapse
|
16
|
Dreon MS, Frassa MV, Ceolín M, Ituarte S, Qiu JW, Sun J, Fernández PE, Heras H. Novel animal defenses against predation: a snail egg neurotoxin combining lectin and pore-forming chains that resembles plant defense and bacteria attack toxins. PLoS One 2013; 8:e63782. [PMID: 23737950 PMCID: PMC3667788 DOI: 10.1371/journal.pone.0063782] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 04/07/2013] [Indexed: 11/19/2022] Open
Abstract
Although most eggs are intensely predated, the aerial egg clutches from the aquatic snail Pomacea canaliculata have only one reported predator due to unparalleled biochemical defenses. These include two storage-proteins: ovorubin that provides a conspicuous (presumably warning) coloration and has antinutritive and antidigestive properties, and PcPV2 a neurotoxin with lethal effect on rodents. We sequenced PcPV2 and studied whether it was able to withstand the gastrointestinal environment and reach circulation of a potential predator. Capacity to resist digestion was assayed using small-angle X-ray scattering (SAXS), fluorescence spectroscopy and simulated gastrointestinal proteolysis. PcPV2 oligomer is antinutritive, withstanding proteinase digestion and displaying structural stability between pH 4.0-10.0. cDNA sequencing and protein domain search showed that its two subunits share homology with membrane attack complex/perforin (MACPF)-like toxins and tachylectin-like lectins, a previously unknown structure that resembles plant Type-2 ribosome-inactivating proteins and bacterial botulinum toxins. The protomer has therefore a novel AB toxin combination of a MACPF-like chain linked by disulfide bonds to a lectin-like chain, indicating a delivery system for the former. This was further supported by observing PcPV2 binding to glycocalix of enterocytes in vivo and in culture, and by its hemaggutinating, but not hemolytic activity, which suggested an interaction with surface oligosaccharides. PcPV2 is able to get into predator's body as evidenced in rats and mice by the presence of circulating antibodies in response to sublethal oral doses. To our knowledge, a lectin-pore-forming toxin has not been reported before, providing the first evidence of a neurotoxic lectin in animals, and a novel function for ancient and widely distributed proteins. The acquisition of this unique neurotoxic/antinutritive/storage protein may confer the eggs a survival advantage, opening new perspectives in the study of the evolution of animal defensive strategies.
Collapse
Affiliation(s)
- Marcos Sebastián Dreon
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Universidad Nacional de La Plata (UNLP) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET CCT-La Plata), La Plata, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Dimitrijevic R, Stojanovic M, Micic M, Dimitrijevic L, Gavrovic-Jankulovic M. Recombinant banana lectin as mucosal immunostimulator. J Funct Foods 2012. [DOI: 10.1016/j.jff.2012.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
18
|
Direct anthelmintic and immunostimulatory effects of oral dosing semi-purified phytohaemagglutinin lectin in sheep infected with Teladorsagia circumcincta and Trichostrongylus colubriformis. Vet Parasitol 2012; 187:267-74. [DOI: 10.1016/j.vetpar.2012.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 12/19/2011] [Accepted: 01/02/2012] [Indexed: 11/20/2022]
|
19
|
Pelosi A, Shepherd R, Walmsley AM. Delivery of plant-made vaccines and therapeutics. Biotechnol Adv 2012; 30:440-8. [DOI: 10.1016/j.biotechadv.2011.07.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 07/14/2011] [Accepted: 07/25/2011] [Indexed: 11/17/2022]
|
20
|
New insights in mucosal vaccine development. Vaccine 2011; 30:142-54. [PMID: 22085556 DOI: 10.1016/j.vaccine.2011.11.003] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 10/25/2011] [Accepted: 11/01/2011] [Indexed: 12/30/2022]
Abstract
Mucosal surfaces are the major entrance for infectious pathogens and therefore mucosal immune responses serve as a first line of defence. Most current immunization procedures are obtained by parenteral injection and only few vaccines are administered by mucosal route, because of its low efficiency. However, targeting of mucosal compartments to induce protective immunity at both mucosal sites and systemic level represents a great challenge. Major efforts are made to develop new mucosal candidate vaccines by selecting appropriate antigens with high immunogenicity, designing new mucosal routes of administration and selecting immune-stimulatory adjuvant molecules. The aim of mucosal vaccines is to induce broad potent protective immunity by specific neutralizing antibodies at mucosal surfaces and by induction of cellular immunity. Moreover, an efficient mucosal vaccine would make immunization procedures easier and be better suited for mass administration. This review focuses on contemporary developments of mucosal vaccination approaches using different routes of administration.
Collapse
|
21
|
Kienle GS, Grugel R, Kiene H. Safety of higher dosages of Viscum album L. in animals and humans--systematic review of immune changes and safety parameters. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2011; 11:72. [PMID: 21871125 PMCID: PMC3180269 DOI: 10.1186/1472-6882-11-72] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 08/28/2011] [Indexed: 12/18/2022]
Abstract
BACKGROUND Viscum album L extracts (VAE, mistletoe) and isolated mistletoe lectins (ML) have immunostimulating properties and a strong dose-dependent cytotoxic activity. They are frequently used in complementary cancer treatment, mainly to improve quality of life, but partly also to influence tumour growth, especially by injecting VAE locally and in high dosage. The question is raised whether these higher dosages can induce any harm or immunosuppressive effects. METHODS Systematic review of all experiments and clinical studies investigating higher dosages of VAE in animals and humans (Viscum album > 1 mg in humans corresponding to > 0.02 mg/kg in animals or ML > 1 ng/kg) and assessing immune parameters or infections or adverse drug reactions. RESULTS 69 clinical studies and 48 animal experiments reported application of higher doses of VAE or ML and had assessed immune changes and/or harm. In these studies, Viscum album was applied in dosages up to 1500 mg in humans and 1400 mg/kg in animals, ML was applied up to 6.4 μg/kg in humans and in animals up to 14 μg/kg subcutaneously, 50 μg/kg nasally and 500 μg/kg orally. A variety of immune parameters showed fluctuating or rising outcomes, but no immunosuppressive effect. Side effects consisted mainly of dose-dependent flu-like symptoms (FLS), fever, local reactions at the injection site and various mild unspecific effects. Occasionally, allergic reactions were reported. After application of high doses of recombinant ML, reversible hepatotoxicity was observed in some cases. CONCLUSIONS Application of higher dosages of VAE or ML is not accompanied by immunosuppression; altogether VAE seems to exhibit low risk but should be monitored by clinicians when applied in high dosages.
Collapse
Affiliation(s)
- Gunver S Kienle
- Institute for Applied Epistemology and Medical Methodology at the University of Witten/Herdecke, Zechenweg 6, 79111 Freiburg, Germany
| | - Renate Grugel
- Institute for Applied Epistemology and Medical Methodology at the University of Witten/Herdecke, Zechenweg 6, 79111 Freiburg, Germany
| | - Helmut Kiene
- Institute for Applied Epistemology and Medical Methodology at the University of Witten/Herdecke, Zechenweg 6, 79111 Freiburg, Germany
| |
Collapse
|
22
|
Jung JH, Kim YH, Song TJ, An H, Kim KD, Kim IB, Yoon TJ, Kim JB. Adjuvant effect of Korean mistletoe lectin on mucosal immunity induction following intranasal immunization with hemagglutinin antigen. Food Sci Biotechnol 2011. [DOI: 10.1007/s10068-011-0089-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
23
|
Rochereau N, Verrier B, Pin JJ, Genin C, Paul S. Phenotypic localization of distinct DC subsets in mouse Peyer Patch. Vaccine 2011; 29:3655-61. [PMID: 21439318 DOI: 10.1016/j.vaccine.2011.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 02/25/2011] [Accepted: 03/05/2011] [Indexed: 12/14/2022]
Abstract
Peyer's patch have been extensively studied as a major inductive site for mucosal immunity within the small intestine. The intestinal mucosa contains numerous dendritic cells, which induce either protective immunity to infectious agents or tolerance to innocuous antigens, including food and commensal bacteria. Although during the past few years, several subsets of human mucosal dendritic cells have been described, a precise characterization of the different mouse mucosal dendritic cells subpopulations remains to be achieved with regard to their phenotype and localization in Peyer's patch. In this report, we have investigated by immunofluorescence on cryosection and by flow cytometry, the phenotype and the localization of dendritic cells into Peyer's patch of C57Bl/6 mouse intestine using dendritic cells markers. Positive and double staining for CD11c and BDCA-2, pDC/IPC, DC-LAMP, DC-SIGN, TLR8 and Langerin have been observed revealing new mouse intestinal DC subsets. This study provides new insight in the understanding of mucosal immune responses induced by natural processes as infections but also new perspectives for the evaluation of oral vaccines.
Collapse
Affiliation(s)
- Nicolas Rochereau
- GIMAP, EA3064, Faculté de médecine, Universités de Lyon, 42023 Saint-Etienne, France
| | | | | | | | | |
Collapse
|
24
|
Wen Z, Yan Z, Hu K, Pang Z, Cheng X, Guo L, Zhang Q, Jiang X, Fang L, Lai R. Odorranalectin-conjugated nanoparticles: preparation, brain delivery and pharmacodynamic study on Parkinson's disease following intranasal administration. J Control Release 2011; 151:131-8. [PMID: 21362449 DOI: 10.1016/j.jconrel.2011.02.022] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 01/12/2011] [Accepted: 02/20/2011] [Indexed: 10/18/2022]
Abstract
Odorranalectin (OL) was recently identified as the smallest lectin with much less immunogenicity than other members of the lectin family. In this study, to improve nose-to-brain drug delivery and reduce the immunogenicity of traditional lectin modified delivery system, OL was conjugated to poly(ethylene glycol)-poly(lactic-co-glycolic acid) (PEG-PLGA) nanoparticles and its biorecognitive activity on nanoparticles was verified by haemagglutination tests. Nose-to-brain delivery characteristic of OL-conjugated nanoparticles (OL-NP) was investigated by in vivo fluorescence imaging technique using DiR as a tracer. Besides, urocortin peptide (UCN), as a macromolecular model drug, was incorporated into nanoparticles and evaluated for its therapeutic efficacy on hemiparkinsonian rats following intranasal administration by rotation behavior test, neurotransmitter determination and tyrosine hydroxylase (TH) test. The results suggested that OL modification increased the brain delivery of nanoparticles and enhanced the therapeutic effects of UCN-loaded nanoparticles on Parkinson's disease. In summary, the OL-NPs could be potentially used as carriers for nose-to-brain drug delivery, especially for macromolecular drugs, in the treatment of CNS disorders.
Collapse
Affiliation(s)
- Ziyi Wen
- Department of Pharmaceutical Science, School of Pharmacy, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Liu Q, Shao X, Chen J, Shen Y, Feng C, Gao X, Zhao Y, Li J, Zhang Q, Jiang X. In vivo toxicity and immunogenicity of wheat germ agglutinin conjugated poly(ethylene glycol)-poly(lactic acid) nanoparticles for intranasal delivery to the brain. Toxicol Appl Pharmacol 2011; 251:79-84. [DOI: 10.1016/j.taap.2010.12.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 11/30/2010] [Accepted: 12/06/2010] [Indexed: 01/29/2023]
|
26
|
Azizi A, Kumar A, Diaz-Mitoma F, Mestecky J. Enhancing oral vaccine potency by targeting intestinal M cells. PLoS Pathog 2010; 6:e1001147. [PMID: 21085599 PMCID: PMC2978714 DOI: 10.1371/journal.ppat.1001147] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The immune system in the gastrointestinal tract plays a crucial role in the control of infection, as it constitutes the first line of defense against mucosal pathogens. The attractive features of oral immunization have led to the exploration of a variety of oral delivery systems. However, none of these oral delivery systems have been applied to existing commercial vaccines. To overcome this, a new generation of oral vaccine delivery systems that target antigens to gut-associated lymphoid tissue is required. One promising approach is to exploit the potential of microfold (M) cells by mimicking the entry of pathogens into these cells. Targeting specific receptors on the apical surface of M cells might enhance the entry of antigens, initiating the immune response and consequently leading to protection against mucosal pathogens. In this article, we briefly review the challenges associated with current oral vaccine delivery systems and discuss strategies that might potentially target mouse and human intestinal M cells.
Collapse
Affiliation(s)
- Ali Azizi
- Infectious Disease and Vaccine Research Center, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.
| | | | | | | |
Collapse
|
27
|
|
28
|
Clement F, Venkatesh YP. Dietary garlic (Allium sativum) lectins, ASA I and ASA II, are highly stable and immunogenic. Int Immunopharmacol 2010; 10:1161-9. [DOI: 10.1016/j.intimp.2010.06.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 06/08/2010] [Accepted: 06/22/2010] [Indexed: 12/16/2022]
|
29
|
Abstract
BACKGROUND Limited accessibility of drugs to the tumor tissues, the requirement of high doses, intolerable cytotoxicity, the development of multiple drug resistance and non-specific targeting are obstacles to the clinical use of cancer drugs and cancer therapy. OBJECTIVE Drug delivery through carrier systems to cancerous tissue is no longer simply wrapping up cancer drugs in a new formulation for different routes of delivery, rather the focus is on targeted cancer therapy. METHODS This review summarizes the exploitation of drug-loaded nanocarrier conjugates with various targeting moieties for the delivery and targeting of anticancer drugs and describes the current status of and challenges in the field of nanocarrier-aided drug delivery and drug targeting. CONCLUSION The discovery of targeting ligand to cancer cells and the development of ligand-targeted therapy will help us to improve therapeutic efficacy and reduce side effects. Unlike other forms of therapy, it will allow us to maintain quality of life for patients, while efficiently attacking the cancer tissue. It indicates that ligands have a pivotal role in cancer cell targeting.
Collapse
Affiliation(s)
- Manasi Das
- Laboratory for Nanomedicine, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Orissa, India
| | | | | |
Collapse
|
30
|
Wu H, Hu K, Jiang X. From nose to brain: understanding transport capacity and transport rate of drugs. Expert Opin Drug Deliv 2009; 5:1159-68. [PMID: 18817519 DOI: 10.1517/17425247.5.10.1159] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The unique relationship between nasal cavity and cranial cavity tissues in anatomy and physiology makes intranasal delivery to the brain feasible. An intranasal delivery provides some drugs with short channels to bypass the blood-brain barrier (BBB), especially for those with fairly low brain concentrations after a routine delivery, thus greatly enhancing the therapeutic effect on brain diseases. In the past two decades, a good number of encouraging outcomes have been reported in the treatment of diseases of the brain or central nervous system (CNS) through nasal administration. In spite of the significant merit of bypassing the BBB, direct nose-to-brain delivery still bears the problems of low efficiency and volume for capacity due to the limited volume of the nasal cavity, the small area ratio of olfactory mucosa to nasal mucosa and the limitations of low dose and short retention time of drug absorption. It is crucial that selective distribution and retention time of drugs or preparations on olfactory mucosa should be enhanced so as to increase the direct delivery efficiency. In this article, we first briefly review the nose-to-brain transport pathways, before detailing the impacts on them, followed by a comprehensive summary of effective methods, including formulation modification, agglutinant-mediated transport and a brain-homing, peptide-mediated delivery based on phage display screening technique, with a view to providing a theoretic reference for elevating the therapeutic effects on brain diseases.
Collapse
Affiliation(s)
- Hongbing Wu
- Fudan University (Fenglin Campus), Department of Pharmaceutics, School of Pharmacy, P.O. Box 130, 138 Yi Xue Yuan Rd, Shanghai 200032, People's Republic of China
| | | | | |
Collapse
|
31
|
Lavelle EC, Grant G, Pfuller U, O'Hagan DT. Immunological Implications of the use of Plant Lectins for Drug and Vaccine Targeting to the Gastrointestinal Tract. J Drug Target 2008; 12:89-95. [PMID: 15203902 DOI: 10.1080/10611860410001693733] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Plant lectins are under consideration as targeting agents to enhance the efficacy of orally administered drugs and vaccines. A significant issue that must be considered is the immunogenicity of these molecules since an immune response to the targeting agent may interfere with its ability to interact with the epithelium. In contrast, the ability of certain lectins to activate the immune system may be exploited in the delivery of vaccines. We previously demonstrated that plant lectins vary widely in their immunogenicity and in particular that mistletoe lectins (ML) I, II and II (MLI, MLII, MLIII) are potent immunogens when administered nasotracheally. Here, we measured immune responses following oral delivery of the MLs and assessed their ability to enhance responses to a co-administered antigen to determine if the molecules possess adjuvant activity. Oral administration of the lectins induced potent lectin-specific systemic and mucosal antibody responses. In addition, each of the three lectins possessed adjuvant activity when delivered orally together with ovalbumin (OVA). The lectins enhanced both serum and mucosal antibody responses to the co-delivered antigen. This shows for the first time that MLI, MLII and MLIII possess adjuvant activity when administered orally and may provide a platform for the generation of effective mucosal adjuvants.
Collapse
Affiliation(s)
- E C Lavelle
- Department of Biochemistry, Trinity College, 2 Dublin, Ireland.
| | | | | | | |
Collapse
|
32
|
Guerrero GG, Feunou F P, Locht C. The coiled-coil N-terminal domain of the heparin-binding haemagglutinin is required for the humoral and cellular immune responses in mice. Mol Immunol 2008; 46:116-24. [PMID: 18801574 DOI: 10.1016/j.molimm.2008.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Accepted: 07/10/2008] [Indexed: 11/17/2022]
Abstract
Heparin-binding haemagglutinin (HBHA) is a 28-kDa mycobacterial adhesin, composed of three functional domains. Previous work has shown that the C-terminal methylated domain is important for adherence, and it is involved in protective T cell immunity in mouse models. However, the role of the coiled-coil N-terminal domain of HBHA in its overall immunogenic capacity remains elusive. Herein, a comparison of the antibody and cellular immune responses after subcutaneous and intranasal immunization of mice with HBHA (native and recombinant) revealed that the methylation pattern is important but not essential for this property. Subcutaneous immunization of mice with a truncated protein, rHBHADeltaC, which lacks the C-terminal methylated domain, was sufficient to trigger humoral and cellular immune responses to HBHA in mice. Altogether we provide evidence that the coiled-coil N-terminal domain is required for HBHA immunogenicity in vivo.
Collapse
|
33
|
Immunity to a self-derived, channel-forming peptide in the respiratory tract. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2007; 15:260-6. [PMID: 18094111 DOI: 10.1128/cvi.00319-07] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The channel-forming peptide NC-1130 was generated based on the amino acid sequence of the M2 segment of the spinal cord alpha-subunit of the glycine receptor and has been proposed as a therapeutic agent for anion channelopathies such as cystic fibrosis. Lysine adduction and amino acid substitutions at positions T19R and S22W of the peptide improved its performance as an ion channel. However, these modifications generated an altered self, potentially making this NC-1130 peptide immunogenic, which could preclude the repeated use of NC-1130 as a therapeutic agent. To measure the ability of NC-1130 to induce an immune response, it was administered nasally with or without cholera toxin (CT). The NC-1130 peptide, when given alone without adjuvant, induced very little peptide-specific immunity based on analyses of peptide-specific antibodies by enzyme-linked immunosorbent assay and enzyme-linked immunospot assay, induction of cytokine production, and delayed-type hypersensitivity (DTH) responses. The administration of NC-1130 with the mucosal adjuvant CT induced peptide-specific immunoglobulin G (IgG) antibodies and DTH responses and a Th2-dominant cytokine response. The coadministration of the strong mucosal adjuvant CT induced a systemic NC-1130-specific IgG response but not a mucosal peptide-specific antibody response. The lack of peptide-specific immunity and specifically mucosal immunity should allow repeated NC-1130 peptide applications to epithelial surfaces to correct anion channelopathies.
Collapse
|
34
|
Kroghsbo S, Madsen C, Poulsen M, Schrøder M, Kvist PH, Taylor M, Gatehouse A, Shu Q, Knudsen I. Immunotoxicological studies of genetically modified rice expressing PHA-E lectin or Bt toxin in Wistar rats. Toxicology 2007; 245:24-34. [PMID: 18215453 DOI: 10.1016/j.tox.2007.12.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 11/30/2007] [Accepted: 12/07/2007] [Indexed: 01/26/2023]
Abstract
As part of the SAFOTEST project the immunmodulating effect of Cry1Ab protein from Bacillus thuringiensis (Bt) and PHA-E lectin from kidney bean (Phaseolus vulgaris erythroagglutinin) was examined in 28- and 90-day feeding studies in Wistar rats. PHA-E lectin was chosen as positive control. Rats were fed control rice, transgenic rice expressing Cry1Ab protein or PHA-E lectin, or transgenic rice spiked with the purified recombinant protein. Total immunoglobulin levels, mitogen-induced cell proliferation, T-dependent antibody response to sheep red blood cells and the antigen-specific antibody response in serum were examined at the end of the studies. A dose-dependent increase in mesenteric lymph node weight and total immunoglobulin A was seen when feeding PHA-E transgenic rice alone or spiked with 0.1% purified PHA-E lectin for 90 days indicating a local effect of PHA-E in the intestine. No adverse effects of Cry1Ab protein were found. An anti-PHA-E and anti-Cry1Ab antibody response was induced both after inhalation (control groups) and after inhalation/ingestion (groups fed recombinant protein alone or together with transgenic rice). In conclusion, only PHA-E lectin was found to have an immunomodulating effect when feeding rats for 90 days with approximately 70 mg PHA-E/kg bodyweight per day. As both PHA-E lectin and Cry1Ab protein were capable of inducing an antigen-specific antibody response it is important to make careful considerations when designing future animal studies to avoid intake of proteins from the other groups by inhalation as well as to examine the sensitization and elicitation potential of 'foreign' proteins before introduction to the world market.
Collapse
Affiliation(s)
- Stine Kroghsbo
- Department of Toxicology and Risk Assessment, National Food Institute, Technical University of Denmark, Mørkhøj Bygade 19, DK-2860 Søborg, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Korean mistletoe lectin (KML-IIU) and its subchains induce nitric oxide (NO) production in murine macrophage cells. J Biomed Sci 2007; 15:197-204. [DOI: 10.1007/s11373-007-9210-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Accepted: 09/12/2007] [Indexed: 11/25/2022] Open
|
36
|
Song SK, Moldoveanu Z, Nguyen HH, Kim EH, Choi KY, Kim JB, Mestecky J. Intranasal immunization with influenza virus and Korean mistletoe lectin C (KML-C) induces heterosubtypic immunity in mice. Vaccine 2007; 25:6359-66. [PMID: 17640781 DOI: 10.1016/j.vaccine.2007.06.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 05/29/2007] [Accepted: 06/10/2007] [Indexed: 11/20/2022]
Abstract
The mucosal adjuvanticity of Korean mistletoe lectin C (KML-C) was investigated in mice intranasally immunized with inactivated influenza virus (H1N1). Mucosal and systemic immune responses were compared to those induced with cholera toxin B subunit (CTB). KML-C increased influenza-specific antibodies with dominant IgG1 subclass in serum, IgG in genital secretions and IgA in saliva, and significantly enhanced influenza-specific lymphocyte proliferation and cytotoxic activity in spleens and in mediastinal lymph nodes. When KML-C was used as a mucosal adjuvant, mice were completely protected from mortality after the challenge with a homologous (H1N1) mouse-adapted influenza virus. After challenge with heterologous (H3N2) influenza virus the level of heterosubtypic immunity in KML-C-treated mice was comparable to that of mice that received CTB as adjuvant. These findings suggest that KML-C may be used as an effective mucosal adjuvant.
Collapse
Affiliation(s)
- Seong Kyu Song
- Institute for Biomedical Research, Han Dong University, Namsong-Ri 3, Buk-Ku, Pohang, Kyungbuk 791-940, South Korea.
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
The oral route is the ideal means of delivering prophylactic and therapeutic vaccines, offering significant advantages over systemic delivery. Most notably, oral delivery is associated with simple administration and improved safety. In addition, unlike systemic immunisation, oral delivery can induce mucosal immune responses. However, the oral route of vaccine delivery is the most difficult because of the numerous barriers posed by the gastrointestinal tract. To facilitate effective immunisation with peptide and protein vaccines, antigens must be protected, uptake enhanced and the innate immune response activated. Numerous delivery systems and adjuvants have been evaluated for oral vaccine delivery, including live vectors, inert particles and bacterial toxins. Although developments in oral vaccines have been disappointing so far, in terms of the generation of products, the availability of a range of novel delivery systems offers much greater hope for the future development of improved oral vaccines.
Collapse
Affiliation(s)
- Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity College, Dublin 2, Ireland.
| | | |
Collapse
|
38
|
Abstract
It is well-established that most pathogens that cause infectious diseases enter the host via mucosal membranes of the respiratory, digestive and genital tracts. Some parenterally administered vaccines induce protection against mucosal pathogens. However, there is increasing evidence that mucosal protection is better afforded by mucosal vaccination, particularly for the induction of memory responses. Mucosal vaccines must pass several difficult hurdles before entering the host and inducing an effective and protective immune response. This review deals with present and past efforts in devising effective mucosal vaccines using delivery systems and immunopotentiating adjuvants for protein-based vaccines. The paper will conclude with the authors' opinion on how the field will or should progress in the future and what will be the required components of ideal future mucosal vaccines that can induce immunological memory.
Collapse
Affiliation(s)
- Michael Vajdy
- Chiron Vaccines, 4560 Horton Street, Emeryville, California 94608, USA.
| | | |
Collapse
|
39
|
Wikstrom ME, Batanero E, Smith M, Thomas JA, von Garnier C, Holt PG, Stumbles PA. Influence of mucosal adjuvants on antigen passage and CD4+ T cell activation during the primary response to airborne allergen. THE JOURNAL OF IMMUNOLOGY 2006; 177:913-24. [PMID: 16818746 DOI: 10.4049/jimmunol.177.2.913] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ag delivery via the nasal route typically induces tolerance or fails to polarize CD4+ T cell responses unless an adjuvant is provided. To better understand this process, we assessed the effects of two mucosal adjuvants, Escherichia coli LPS and cholera toxin (CT), on Ag passage and T cell activation in the draining lymph nodes (DLN) of BALB/c mice following per nasal administration of the model protein allergen, OVA. We found a range of cell types acquired small amounts of fluorescent OVA in the DLN 4 h after per nasal administration. However, this early uptake was eclipsed by a wave of OVA+CD8alpha(low) dendritic cells that accumulated in the DLN over the next 20 h to become the dominant OVA-processing and -presenting population. Both LPS and CT stimulated increases in CD80 and CD86 expression on OVA+CD8alpha(low) DC. LPS also increased the number of OVA+CD8alpha(low) dendritic cells accumulating in the DLN. When the primary T cell response was examined after adoptive transfer of CD4+ T cells from DO11.10 mice, CT and LPS stimulated surprisingly similar effects on T cell activation and proliferation, IL-4 and IFN-gamma priming, and memory T cell production. Despite these similarities, T cell recipients immunized with CT, but not LPS, developed lung eosinophilia upon secondary OVA challenge. Thus, we found no bias within the DLN in Ag handling or the primary T cell response associated with the eventual Th2 polarization induced by CT, and suggest that additional tissue-specific factors influence the development of allergic disease in the airways.
Collapse
Affiliation(s)
- Matthew E Wikstrom
- Division of Cell Biology, Telethon Institute for Child Health Research, West Perth, WA 6872, Australia.
| | | | | | | | | | | | | |
Collapse
|
40
|
Lavelle EC. Lectins and microparticles for enhanced oral vaccination. Methods 2006; 38:84-9. [PMID: 16406570 DOI: 10.1016/j.ymeth.2005.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Accepted: 09/16/2005] [Indexed: 12/25/2022] Open
Abstract
Oral administration is an attractive means of vaccination since it alleviates the need for injection and trained personnel. Furthermore, effective oral vaccination induces immune responses locally in the gastrointestinal tract, the site of colonisation and infection by many pathogens. However, since most proteins and peptides are degraded in the gut, do not interact specifically with the epithelium, or induce strong immune responses, this route of delivery is generally inefficient. Two of the most promising potential approaches are the use of lectins to target vaccines to the epithelium and the association of vaccines with biodegradable microparticles. This paper describes methods to assess the stability of vaccines and targeting molecules in the gastrointestinal tract, interaction with the mucosa, uptake into internal organs and induction of mucosal antibody responses.
Collapse
Affiliation(s)
- E C Lavelle
- Adjuvant Research Group, Department of Biochemistry, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
41
|
|
42
|
Abstract
Lectins are carbohydrate binding (glyco)proteins which are ubiquitous in nature. In plants, they are distributed in various families and hence ingested daily in appreciable amounts by both humans and animals. One of the most nutritionally important features of plant lectins is their ability to survive digestion by the gastrointestinal tract of consumers. This allows the lectins to bind to membrane glycosyl groups of the cells lining the digestive tract. As a result of this interaction a series of harmful local and systemic reactions are triggered placing this class of molecules as antinutritive and/or toxic substances. Locally, they can affect the turnover and loss of gut epithelial cells, damage the luminal membranes of the epithelium, interfere with nutrient digestion and absorption, stimulate shifts in the bacterial flora and modulate the immune state of the digestive tract. Systemically, they can disrupt lipid, carbohydrate and protein metabolism, promote enlargement and/or atrophy of key internal organs and tissues and alter the hormonal and immunological status. At high intakes, lectins can seriously threaten the growth and health of consuming animals. They are also detrimental to numerous insect pests of crop plants although less is presently known about their insecticidal mechanisms of action. This current review surveys the recent knowledge on the antinutritional/toxic effects of plant lectins on higher animals and insects.
Collapse
Affiliation(s)
- Ilka M Vasconcelos
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Caixa Postal 6020, Campus do Pici, CEP 60451-970, Fortaleza, CE, Brazil.
| | | |
Collapse
|
43
|
Gabor F, Bogner E, Weissenboeck A, Wirth M. The lectin-cell interaction and its implications to intestinal lectin-mediated drug delivery. Adv Drug Deliv Rev 2004; 56:459-80. [PMID: 14969753 DOI: 10.1016/j.addr.2003.10.015] [Citation(s) in RCA: 181] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2003] [Accepted: 10/14/2003] [Indexed: 01/09/2023]
Abstract
Based on the fact that oligosaccharides encode biological information, the biorecognition between lectinised drug delivery systems and glycosylated structures in the intestine can be exploited for improved peroral therapy. Basic research revealed that some lectins can mediate mucoadhesion, cytoadhesion, and cytoinvasion of drugs. Entering the vesicular pathway by receptor mediated endocytosis, part of the conjugated drug is accumulated within the lysosomes. Additionally, part of the drug is supposed to be transported across the epithelium. Moreover, factors probably adversely influencing feasibility of the concept such as toxicity, immunogenicity, and intestinal stability of plant lectins are discussed. As exemplified by lectin-grafted prodrug and carrier systems, this strategy is expected to improve absorption and probably bioavailability of poorly absorbable drugs, peptides and proteins as well as therapeutic DNA.
Collapse
Affiliation(s)
- Franz Gabor
- Institute of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria.
| | | | | | | |
Collapse
|
44
|
Walter F, Schöll I, Untersmayr E, Ellinger A, Boltz-Nitulescu G, Scheiner O, Gabor F, Jensen-Jarolim E. Functionalisation of allergen-loaded microspheres with wheat germ agglutinin for targeting enterocytes. Biochem Biophys Res Commun 2004; 315:281-7. [PMID: 14766205 DOI: 10.1016/j.bbrc.2004.01.057] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2003] [Indexed: 11/17/2022]
Abstract
In this study, we constructed particles applicable for oral immunotherapy of type I allergy by protecting allergens from digestion and supporting intestinal antigen uptake. Therefore, birch-pollen allergens were entrapped in poly(d,l-lactic-co-glycolic acid) microspheres by spray-drying rendering microspheres with a main population of 1-3microm. Microspheres were further coated with wheat germ agglutinin (WGA) to target enterocytes. Coating with WGA did not alter the surface characteristics of the microspheres as demonstrated in scanning electron microscopy. Binding of WGA was specific and could be inhibited by chitotriose to 14.7+/-6.9%. Comparable amounts of allergen were released from both particle-types with 46.3+/-1.7% and 44.5+/-2.6% during 21 days. Simulating gastric digestion in vitro, antigenicity of allergens entrapped in WGA-microspheres was preserved to 59.8+/-1.5% even after 2h. Feedings of BALB/c mice with WGA-microspheres induced higher levels of allergen-specific IgG-levels than gavages of uncoated microparticles or naked protein. Thus, we conclude that WGA-microspheres are suitable vehicles for oral delivery and mucosal targeting due to lectin-mediated bioadhesion.
Collapse
Affiliation(s)
- Franziska Walter
- Department of Pathophysiology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna A-1090, Austria
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Lavelle EC, Jarnicki A, McNeela E, Armstrong ME, Higgins SC, Leavy O, Mills KHG. Effects of cholera toxin on innate and adaptive immunity and its application as an immunomodulatory agent. J Leukoc Biol 2004; 75:756-63. [PMID: 14704372 DOI: 10.1189/jlb.1103534] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Cholera toxin (CT) is a potent vaccine adjuvant when administered via parenteral, mucosal, or transcutaneous routes. It also inhibits innate inflammatory responses induced by pathogen-derived molecules, such as lipopolysaccharide (LPS). We demonstrated previously that CT promotes the induction of regulatory type 1 T cells (Tr1) as well as T helper type 2 cells (Th2). T cells from mice immunized with antigen in the presence of CT produced high levels of interleukin (IL)-10 and IL-5 and low levels of IL-4 and interferon-gamma (IFN-gamma). Here, we demonstrate that immunization with antigen in the presence of CT induced a population of antigen-specific CD4(+) T cells that produced IL-10 in the absence of IL-4, in addition to cells that coexpressed IL-4 and IL-10 or produced IL-4 only. CT-generated Tr1 cells inhibited antigen-specific proliferation as well as IFN-gamma production by Th1 cells, and this suppression was cell contact-independent. It is interesting that coincubation with Th1 cells significantly enhanced IL-10 production by the Tr1 cells. As IL-10 can promote the differentiation of Tr1 cells, we investigated cytokine production by dendritic cells (DC) following exposure to CT. Previous data showed that CT can modulate the expression of costimulatory molecules and inhibit the production of chemokines and cytokines, including IL-12 and tumor necrosis factor alpha and enhance IL-10 production. Here, we show that CT synergizes with LPS to induce IL-6 and IL-1beta in addition to IL-10 production by immature DC. Therefore, CT may promote the induction of Th2 and Tr1 cells in part via selective modulation of DC cytokine production and costimulatory molecule expression.
Collapse
Affiliation(s)
- Ed C Lavelle
- Department of Biochemistry, Trinity College, Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Developing efficient adjuvants for human vaccines that elicit broad and sustained immune responses at systemic or mucosal levels remains a formidable challenge for the vaccine industry. Conventional approaches in the past have been largely empirical and--at best--partially successful. Importantly, recent advances in our understanding of the immune system, most particularly with respect to early proinflammatory signals, are leading to the identification of new biological targets for vaccine adjuvants. This review covers both the current status of adjuvant testing in humans, the residual needs for vaccines in development, and the emerging immunological foundations for adjuvant design. A better understanding of the biology of toll-like receptors, non-conventional T cell subpopulations, T and B cell memory, regulatory T cells, and mucosal immunity has profound implications for a modern approach to adjuvant screening and development. The future lies in the high throughput screening of synthetic chemical entities targeting well-characterized biological molecules. Used alone or in combination, such synthetic adjuvants will allow stimulation or modulation in a safe and efficient manner of strong effector, regulatory and memory immune mechanisms.
Collapse
Affiliation(s)
- Nicolas Burdin
- Aventis Pasteur, Research and Development, Campus Mérieux, Marcy l'Etoile, France
| | | | | |
Collapse
|
47
|
Abstract
Current efforts to improve the effectiveness of microparticle vaccines include incorporating biomimetic features into the particles. Many pathogens use surface molecules to target specific cell types in the gut for host invasion. This observation has inspired efforts to chemically conjugate cell-type targeting ligands to the surfaces of microparticles in order to increase the efficiency of uptake, and therefore the effectiveness, of orally administered microparticles. Bio-mimicry is not limited to the exterior surface of the microparticles. Anti-idiotypic antibodies, cytokines or other biological modifiers can be encapsulated for delivery to sites of interest as vaccines or other therapeutics. Direct mucosal delivery of microparticle vaccines or immunomodulatory agents may profoundly enhance mucosal and systemic immune responses compared to other delivery routes.
Collapse
Affiliation(s)
- Mark E Keegan
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | | |
Collapse
|
48
|
Camposampiero D, Tiso R, Zanetti E, Ruzza A, Bruni A, Ponzin D. Cornea preservation in culture with bovine serum or chicken ovalbumin. Cornea 2003; 22:254-8. [PMID: 12658094 DOI: 10.1097/00003226-200304000-00015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE To explore the possibility of replacing bovine serum with chicken ovalbumin in cornea preservation. METHODS Twenty-one pairs of corneas were cultivated at 31 degrees C in a medium containing either 2% (vol/vol) of newborn calf serum (one cornea) or 2 mg/mL of chicken ovalbumin (the contralateral cornea). The evaluation of corneal endothelium was performed after 7 days (one pair) and between 11 and 37 days (20 pairs). Evaluation included the number and the morphology of endothelial cells, their viability, and the pattern of swelling of the intercellular borders. RESULTS A preliminary test showed that the incubation of a cornea for 7 days with ovalbumin did not cause acute toxic effects. Subsequent experiments showed that this protein was as efficient as bovine serum in preserving the viability and the density of endothelial cells for long periods of incubation. After a mean of 21 +/- 10 days of culture, the number of endothelial cells was 2,455 +/- 230/mm2 in the serum group and 2,430 +/- 181/mm2 in the ovalbumin group. During the longest incubation period, irregular swelling of cell borders and other signs of endothelial degeneration were occasionally observed. CONCLUSION Replacing serum with chicken ovalbumin resulted in the efficient preservation of corneal endothelium for at least 3 weeks of culture. These data suggest that ovalbumin may be an effective basic component in the design of a serum-free culture medium.
Collapse
|
49
|
Kjaer TMR, Frøkiaer H. Modulation of ovomucoid-specific oral tolerance in mice fed plant extracts containing lectins. Br J Nutr 2002; 88:671-80. [PMID: 12493089 DOI: 10.1079/bjn2002736] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We investigated the effect of feeding extracts of four different legumes (red kidney bean (Phaseolus vulgaris), peanut (Arachis hypogaea), soyabean (Glycine max) and pea (Pisum sativum) on the specific immune response against a food protein. Mice were fed ovomucoid and the specific immune response was evaluated. Ovomucoid fed alone resulted in oral tolerance induction measured as both a reduced ovomucoid-specific spleen cell proliferation and antibody response. Feeding kidney-bean extract prevented induction of oral tolerance to ovomucoid measured as spleen cell proliferation in vitro. Pure kidney-bean lectin also prevented oral tolerance induction, suggesting that lectin in the kidney-bean extract caused inhibition of oral tolerance. Parenteral administration (intravenous and intraperitoneal) of pure kidney-bean lectin had no significant influence on oral tolerance induction. Soyabean extract also influenced the immune response against ovomucoid; however, this was not as pronounced as for kidney bean and was only significant (P<0.001) for the antibody response. No effect was observed when pea extract was fed and peanut extract had a non-significant effect on induction of oral tolerance and on the general immune response. Plasma antibodies against kidney-bean lectin, but not against the three other legume lectins, were detected. Our current findings show that other dietary components can influence the specific immune response against food proteins. Various dietary components may thus contribute to the onset of adverse immunological responses.
Collapse
Affiliation(s)
- Tanja M R Kjaer
- BioCentrum-DTU, Biochemistry and Nutrition, Technical University of Denmark, Lyngby, Denmark
| | | |
Collapse
|
50
|
Lavelle EC, Grant G, Pusztai A, Pfüller U, Leavy O, McNeela E, Mills KHG, O'Hagan DT. Mistletoe lectins enhance immune responses to intranasally co-administered herpes simplex virus glycoprotein D2. Immunology 2002; 107:268-74. [PMID: 12383207 PMCID: PMC1782787 DOI: 10.1046/j.1365-2567.2002.01492.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mucosal adjuvant properties of the three type 2 ribosome-inactivating proteins (RIPs) from the European mistletoe, Viscum album L., were investigated. Mistletoe lectins were compared with cholera toxin (CT) as adjuvants when delivered nasotracheally together with herpes simplex virus glycoprotein D2 (gD2). All three mistletoe lectins (MLI, MLII, MLIII) were potent mucosal adjuvants. Co-administration of MLI, MLII or MLIII with gD2 led to significantly higher levels of gD2-specific mucosal immunoglobulin A (IgA) and systemic immunoglobulin G (IgG) antibody than when the antigen was delivered alone. The levels of antibodies induced were similar to those generated in mice immunized with gD2 and the potent mucosal adjuvant CT. Administration of ML1 with gD2 enhanced the antigen-specific splenic T-cell proliferative response. Interleukin-5 (IL-5), but not interferon-gamma (IFN-gamma), was detected in supernatants from splenocytes stimulated in vitro with gD2. This indicates that MLI enhanced type 2 T-helper cell (Th2) responses to the bystander antigen, gD2. Analysis of the gD2- and lectin-specific IgG subclass titres in mice immunized with gD2 and MLI, MLII or MLIII revealed a high ratio of IgG1 : IgG2a, which is compatible with the selective induction of Th2-type immune responses.
Collapse
Affiliation(s)
- E C Lavelle
- Rowett Research Institute, Bucksburn, Aberdeen, Scotland, UK.
| | | | | | | | | | | | | | | |
Collapse
|