Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Transl Res ; 263: 45-52, 2024 01.
Article in English | MEDLINE | ID: mdl-37678755

ABSTRACT

Cyclic nucleotide elevation in intestinal epithelial cells is the key pathology causing intestinal fluid loss in secretory diarrheas such as cholera. Current secretory diarrhea treatment is primarily supportive, and oral rehydration solution is the mainstay of cholera treatment. There is an unmet need for safe, simple and effective diarrhea treatments. By promoting cAMP hydrolysis, extracellular calcium-sensing receptor (CaSR) is a regulator of intestinal fluid transport. We studied the antidiarrheal mechanisms of FDA-approved CaSR activator cinacalcet and tested its efficacy in clinically relevant human cell, mouse and intestinal organoid models of secretory diarrhea. By using selective inhibitors, we found that cAMP agonists-induced secretory short-circuit currents (Isc) in human intestinal T84 cells are mediated by collective actions of apical membrane cystic fibrosis transmembrane conductance regulator (CFTR) and Clc-2 Cl- channels, and basolateral membrane K+ channels. 30 µM cinacalcet pretreatment inhibited all 3 components of forskolin and cholera toxin-induced secretory Isc by ∼75%. In mouse jejunal mucosa, cinacalcet inhibited forskolin-induced secretory Isc by ∼60% in wild type mice, with no antisecretory effect in intestinal epithelia-specific Casr knockout mice (Casr-flox; Vil1-cre). In suckling mouse model of cholera induced by oral cholera toxin, single dose (30 mg/kg) oral cinacalcet treatment reduced intestinal fluid accumulation by ∼55% at 20 hours. Lastly, cinacalcet inhibited forskolin-induced secretory Isc by ∼75% in human colonic and ileal organoids. Our findings suggest that CaSR activator cinacalcet has antidiarrheal efficacy in distinct human cell, organoid and mouse models of secretory diarrhea. Considering its excellent clinical safety profile, cinacalcet can be repurposed as a treatment for cyclic nucleotide-mediated secretory diarrheas including cholera.


Subject(s)
Antidiarrheals , Cholera , Mice , Humans , Animals , Antidiarrheals/metabolism , Antidiarrheals/pharmacology , Antidiarrheals/therapeutic use , Cholera/drug therapy , Cholera/metabolism , Cholera/pathology , Cholera Toxin/metabolism , Cholera Toxin/pharmacology , Cholera Toxin/therapeutic use , Cinacalcet/pharmacology , Cinacalcet/therapeutic use , Cinacalcet/metabolism , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/therapeutic use , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/pharmacology , Nucleotides, Cyclic/therapeutic use , Colforsin/metabolism , Colforsin/pharmacology , Colforsin/therapeutic use , Diarrhea/drug therapy , Diarrhea/metabolism , Intestinal Mucosa/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/therapeutic use , Mice, Knockout
2.
Molecules ; 28(24)2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38138443

ABSTRACT

Cholera is an exceptionally aggressive infectious disease characterized by the potential to induce acute, copious, watery diarrhea of considerable severity and renal inflammation. Diabetic nephropathy is a serious complication of diabetes mellitus that can lead to kidney failure through inflammation; thus, anti-inflammatory agents are promising therapies for diabetic nephropathy. Previous studies have shown that the essential oil of Zanthoxylum myriacanthum var. pubescens Huang, Maqian essential oil (MQEO), exhibits potent antibacterial, anti-inflammatory, and renoprotective activities in diabetic mice and has emerged as a potential therapeutic drug for the treatment of diabetic nephropathy complications. Therefore, the present study was carried out to screen the potential inhibition of cholera toxin and the diabetic renoprotective activity of MQEO through computational approaches. Twelve chemical constituents derived from MQEO were docked with cholera toxin and the target proteins involved in diabetic nephropathy, namely, TXNIP, Nrf2, and DPP IV, and, subsequently, the predictions of molecular dynamic simulations, the drug-likeness properties, and the ADMET properties were performed. α-terpineol showed high binding affinities toward the cholera toxin protein. For TXNIP, among all the chemical constituents, α-phellandrene and p-cymene showed strong binding affinities with the TXNIP protein and displayed relatively stable flexibility at the hinge regions of the protein, favorable physicochemical properties in the absence of hepatotoxicity, and low cytotoxicity. For Nrf2, α-terpineol exhibited the highest binding affinity and formed a very stable complex with Nrf2, which displayed high pharmacokinetic properties. All compounds had low free-binding energies when docked with the DPP IV protein, which suggests potent biological activity. In conclusion, based on a computational approach, our findings reveal that MQEO constituents have inhibitory activity against cholera toxin and are promising therapeutic agents for suppressing diabetic inflammation and for the treatment of diabetic nephropathy complications.


Subject(s)
Cholera , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Oils, Volatile , Mice , Animals , Diabetic Nephropathies/metabolism , Oils, Volatile/pharmacology , Oils, Volatile/therapeutic use , Cholera Toxin/chemistry , Cholera Toxin/metabolism , Cholera Toxin/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Cholera/complications , Cholera/drug therapy , Molecular Dynamics Simulation , NF-E2-Related Factor 2/metabolism , Inflammation/drug therapy , Anti-Inflammatory Agents/pharmacology
3.
J Appl Microbiol ; 133(6): 3605-3616, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36000378

ABSTRACT

AIMS: The present study aimed to document the comparative analysis of differential hypervirulent features of Vibrio cholerae O1 strains isolated during 2018 from cholera endemic regions in Gujarat and Maharashtra (Western India) and West Bengal (Eastern India). METHODS AND RESULTS: A total of 87 V. cholerae O1 clinical strains from Western India and 48 from Eastern India were analysed for a number of biotypic and genotypic features followed by antimicrobial resistance (AMR) profile. A novel polymerase chain reaction was designed to detect a large fragment deletion in the Vibrio seventh pandemic island II (VSP-II) genomic region, which is a significant genetic feature of the V. cholerae strains that have caused Yemen cholera outbreak. All the strains from Western India belong to the Ogawa serotype, polymyxin B-sensitive, hemolytic, had a deletion in VSP-II (VSP-IIC) region and carried Haitian genetic alleles of ctxB, tcpA and rtxA. Conversely, 14.6% (7/48) of the strains from Eastern India belonged to the Inaba serotype, polymyxin B-resistant, nonhemolytic, harboured VSP-II other than VSP-IIC type, classical ctxB, Haitian tcpA and El Tor rtxA alleles. Resistance to tetracycline and chloramphenicol has been observed in strains from both regions. CONCLUSIONS: This study showed hypervirulent, polymyxin B-sensitive epidemic causing strains in India along with the strains with polymyxin B-resistant and nonhemolytic traits that may spread and cause serious disease outcomes in future. SIGNIFICANCE AND IMPACT OF THE STUDY: The outcomes of this study can help to improve the understanding of the hyperpathogenic property of recently circulating pandemic Vibrio cholerae strains in India. Special attention is also needed for the monitoring of AMR surveillance because V. cholerae strains are losing susceptibility to many antibiotics used as a second line of defence in the treatment of cholera.


Subject(s)
Cholera , Vibrio cholerae O1 , Humans , Vibrio cholerae O1/genetics , Cholera/epidemiology , Cholera/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Polymyxin B/pharmacology , Haiti , Drug Resistance, Bacterial/genetics , India/epidemiology , Genotype , Disease Outbreaks , Cholera Toxin/genetics , Cholera Toxin/therapeutic use
4.
Toxins (Basel) ; 14(3)2022 03 18.
Article in English | MEDLINE | ID: mdl-35324722

ABSTRACT

Vibrio cholerae uses cholera toxin (CT) to cause cholera, a severe diarrheal disease in humans that can lead to death within hours of the onset of symptoms. The catalytic activity of CT in target epithelial cells increases cellular levels of 3',5'-cyclic AMP (cAMP), leading to the activation of the cystic fibrosis transmembrane conductance regulator (CFTR), an apical ion channel that transports chloride out of epithelial cells, resulting in an electrolyte imbalance in the intestinal lumen and massive water loss. Here we report that when administered perorally, benzopyrimido-pyrrolo-oxazinedione, (R)-BPO-27), a potent small molecule inhibitor of CFTR, blocked disease symptoms in a mouse model for acute diarrhea caused by toxigenic V. cholerae. We show that both (R)-BPO-27 and its racemic mixture, (R/S)-BPO-27, are able to protect mice from CT-dependent diarrheal disease and death. Furthermore, we show that, consistent with the ability of the compound to block the secretory diarrhea induced by CT, BPO-27 has a measurable effect on suppressing the gut replication and survival of V. cholerae, including a 2010 isolate from Haiti that is representative of the most predominant 'variant strains' that are causing epidemic and pandemic cholera worldwide. Our results suggest that BPO-27 should advance to human Phase I studies that could further address its safety and efficacy as therapeutic or preventative drug intervention for diarrheal syndromes, including cholera, that are mediated by CFTR channel activation.


Subject(s)
Cholera , Vibrio cholerae , Animals , Cholera/drug therapy , Cholera Toxin/therapeutic use , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/therapeutic use , Diarrhea/drug therapy , Mice , Morbidity , Vibrio cholerae/metabolism
5.
J Control Release ; 343: 551-563, 2022 03.
Article in English | MEDLINE | ID: mdl-35124128

ABSTRACT

Macromolecular drugs are widely thought to be one of the most promising fields, but there are still many problems, especially with regard to drug delivery. Drug delivery systems are focused on loading efficiency without loss of activity, effective cellular internalization, anti-degradation, target ability, etc. New directions for macromolecular drugs delivery systems are not only to retain the activity of drugs, but bring new bioactivity to carry out dual benefits. Cholera toxin (CT) from Vibrio cholerae is one of such delivery systems and plays a potential role in delivering macromolecular drugs. After released from V. cholerae in the intestine, the B subunit of CT binds to the ganglioside GM1 on intestinal cells, and then the toxin gains access into the intestine. CT has potential as a "vaccine adjuvant-delivery system" (VADS) and is able to bring antigens and serve as adjuvants to induce specific immunity. In addition, it has been well used in the field of mucosal drug delivery and neural targeting. However, native CT is toxic, which restricts its practical application. There are several CT-based proteins with reduced virulence and reserved or even enhanced adjuvant activity under research. In this review, we comprehensively summarize the preparation strategy, advantages, applications and corresponding deficiencies of CT-based proteins. CT is focused on a delivery system when delivering macromolecular cargos such as active protein/peptide and antigen/antigen peptide. CT-based drug delivery system deserves further study due to their superiority.


Subject(s)
Cholera Toxin , Drug Delivery Systems , Adjuvants, Immunologic/metabolism , Cholera Toxin/metabolism , Cholera Toxin/therapeutic use , Drug Delivery Systems/methods , G(M1) Ganglioside/metabolism , Mucous Membrane
6.
Toxins (Basel) ; 13(6)2021 05 26.
Article in English | MEDLINE | ID: mdl-34073185

ABSTRACT

The B subunit pentamer verotoxin (VT aka Shiga toxin-Stx) binding to its cellular glycosphingolipid (GSL) receptor, globotriaosyl ceramide (Gb3) mediates internalization and the subsequent receptor mediated retrograde intracellular traffic of the AB5 subunit holotoxin to the endoplasmic reticulum. Subunit separation and cytosolic A subunit transit via the ER retrotranslocon as a misfolded protein mimic, then inhibits protein synthesis to kill cells, which can cause hemolytic uremic syndrome clinically. This represents one of the most studied systems of prokaryotic hijacking of eukaryotic biology. Similarly, the interaction of cholera AB5 toxin with its GSL receptor, GM1 ganglioside, is the key component of the gastrointestinal pathogenesis of cholera and follows the same retrograde transport pathway for A subunit cytosol access. Although both VT and CT are the cause of major pathology worldwide, the toxin-receptor interaction is itself being manipulated to generate new approaches to control, rather than cause, disease. This arena comprises two areas: anti neoplasia, and protein misfolding diseases. CT/CTB subunit immunomodulatory function and anti-cancer toxin immunoconjugates will not be considered here. In the verotoxin case, it is clear that Gb3 (and VT targeting) is upregulated in many human cancers and that there is a relationship between GSL expression and cancer drug resistance. While both verotoxin and cholera toxin similarly hijack the intracellular ERAD quality control system of nascent protein folding, the more widespread cell expression of GM1 makes cholera the toxin of choice as the means to more widely utilise ERAD targeting to ameliorate genetic diseases of protein misfolding. Gb3 is primarily expressed in human renal tissue. Glomerular endothelial cells are the primary VT target but Gb3 is expressed in other endothelial beds, notably brain endothelial cells which can mediate the encephalopathy primarily associated with VT2-producing E. coli infection. The Gb3 levels can be regulated by cytokines released during EHEC infection, which complicate pathogenesis. Significantly Gb3 is upregulated in the neovasculature of many tumours, irrespective of tumour Gb3 status. Gb3 is markedly increased in pancreatic, ovarian, breast, testicular, renal, astrocytic, gastric, colorectal, cervical, sarcoma and meningeal cancer relative to the normal tissue. VT has been shown to be effective in mouse xenograft models of renal, astrocytoma, ovarian, colorectal, meningioma, and breast cancer. These studies are herein reviewed. Both CT and VT (and several other bacterial toxins) access the cell cytosol via cell surface ->ER transport. Once in the ER they interface with the protein folding homeostatic quality control pathway of the cell -ERAD, (ER associated degradation), which ensures that only correctly folded nascent proteins are allowed to progress to their cellular destinations. Misfolded proteins are translocated through the ER membrane and degraded by cytosolic proteosome. VT and CT A subunits have a C terminal misfolded protein mimic sequence to hijack this transporter to enter the cytosol. This interface between exogenous toxin and genetically encoded endogenous mutant misfolded proteins, provides a new therapeutic basis for the treatment of such genetic diseases, e.g., Cystic fibrosis, Gaucher disease, Krabbe disease, Fabry disease, Tay-Sachs disease and many more. Studies showing the efficacy of this approach in animal models of such diseases are presented.


Subject(s)
Bacterial Toxins/therapeutic use , Animals , Cholera Toxin/therapeutic use , Endoplasmic Reticulum/metabolism , Gaucher Disease/drug therapy , Humans , Protein Subunits/therapeutic use , Proteostasis Deficiencies/drug therapy , Shiga Toxins/therapeutic use , Trihexosylceramides/metabolism
7.
Toxins (Basel) ; 11(12)2019 11 20.
Article in English | MEDLINE | ID: mdl-31756977

ABSTRACT

Cholera toxin B subunit (CTB), a non-toxic homopentameric component of Vibrio cholerae holotoxin, is an oral cholera vaccine antigen that induces an anti-toxin antibody response. Recently, we demonstrated that a recombinant CTB variant with a Lys-Asp-Glu-Leu (KDEL) endoplasmic reticulum retention motif (CTB-KDEL) exhibits colon mucosal healing effects that have therapeutic implications for inflammatory bowel disease (IBD). Herein, we investigated the feasibility of CTB-KDEL for the treatment of chronic colitis. We found that weekly oral administration of CTB-KDEL, dosed before or after the onset of chronic colitis, induced by repeated dextran sodium sulfate (DSS) exposure, could significantly reduce disease activity index scores, intestinal permeability, inflammation, and histological signs of chronicity. To address the consequences of immunogenicity, mice (C57BL/6 or C3H/HeJ strains) were pre-exposed to CTB-KDEL then subjected to DSS colitis and CTB-KDEL treatment. While the pre-dosing of CTB-KDEL elicited high-titer anti-drug antibodies (ADAs) of the immunoglobin A (IgA) isotype in the intestine of C57BL/6 mice, the therapeutic effects of CTB-KDEL were similar to those observed in C3H/HeJ mice, which showed minimal ADAs under the same experimental conditions. Thus, the immunogenicity of CTB-KDEL does not seem to impede the protein's mucosal healing efficacy. These results support the development of CTB-KDEL for IBD therapy.


Subject(s)
Cholera Toxin/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Oligopeptides , Protein Sorting Signals , Animals , Chronic Disease , Cytokines/metabolism , Dextran Sulfate , Female , Immunoglobulin A/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Recombinant Proteins/therapeutic use
8.
Toxins (Basel) ; 9(12)2017 11 23.
Article in English | MEDLINE | ID: mdl-29168738

ABSTRACT

Cholera toxin B subunit (CTB) is a mucosal immunomodulatory protein that induces robust mucosal and systemic antibody responses. This well-known biological activity has been exploited in cholera prevention (as a component of Dukoral® vaccine) and vaccine development for decades. On the other hand, several studies have investigated CTB's immunotherapeutic potential in the treatment of inflammatory diseases such as Crohn's disease and asthma. Furthermore, we recently found that a variant of CTB could induce colon epithelial wound healing in mouse colitis models. This review summarizes the possible mechanisms behind CTB's anti-inflammatory activity and discuss how the protein could impact mucosal inflammatory disease treatment.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cholera Toxin/therapeutic use , Immunologic Factors/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Cholera Toxin/chemistry , Epithelial Cells/drug effects , Humans , Immunologic Factors/chemistry , Intestinal Mucosa/cytology , Molecular Structure
9.
J Neuroinflammation ; 13(1): 147, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27296014

ABSTRACT

BACKGROUND: Cholera toxin B subunit (CTB) has multifaceted immunoregulatory functions. Immunity plays an important role in the mechanism of stroke. However, little is known about whether CTB is beneficial for stroke. METHODS: CTB was administered intraperitoneally after ischemia to rats subjected to transient focal ischemia. Infarct volumes, body weight loss, and neurologic deficits were measured. Cytokines, microglia/macrophage activation, and transcriptional factors in the ischemic brain were tested. The mRNA expressions of IL-1ß and TNF-α were tested in the microglia/macrophage isolated from the ischemic hemisphere. γδT cells, IL-17-producing γδT cells, Th17 cells, and regulatory T (Treg) cells in the ischemic brain were tested. γδT cells and Treg cells in the peripheral blood were also evaluated. RESULTS: CTB reduced infarct volumes, neurologic deficits, and body weight loss after ischemia. At 24 h after ischemia, CTB downregulated the levels of IL-1ß, TNF-α, NF-kB p65, phosphorylated-ERK1/2, and microglia/macrophage activation and suppressed NF-kB binding activity, but did not affect the level of ERK1/2. The mRNA expressions of IL-1ß and TNF-α in the microglia/macrophage isolated from the ischemic hemisphere were suppressed after CTB therapy. In the ischemic hemisphere, CTB treatment reduced the levels of γδT cells, IL-17-producing γδT cells, and IL-17 at both 24 and 72 h after ischemia, while Th17 cells were not affected. After CTB treatment, the levels of Treg cells, TGF-ß, and IL-10 remained unchanged at 24 h and upregulated at 72 h after ischemia. Inactivation of Treg cells using anti-CD25 attenuated the increase of TGF-ß and IL-10 induced by CTB at 72 h after ischemia. In the peripheral blood, CTB increased Treg cells and suppressed γδT cells at 24 h after ischemia. And then at 72 h after ischemia, it increased Treg cells but did not impact γδT cells. CTB had no effect on cytokines, transcription factors, infiltrating γδT cells, and Treg cells in the brain of shams. In the peripheral blood of shams, CTB increased Treg cells at both 24 and 72 h, while it did not affect γδT cells. CONCLUSIONS: CTB decreased neurologic impairment and tissue injury after cerebral ischemia via its immunomodulatory functions, including inhibiting microglia/macrophage activation, suppressing γδT cells, and inducing production of Treg cells, thus regulating the secretion of related cytokines. Suppression of NF-kB and ERK1/2 pathways is involved in the neuroprotective mechanism of CTB.


Subject(s)
Cholera Toxin/therapeutic use , Cytokines/metabolism , Encephalitis/drug therapy , Encephalitis/etiology , Infarction, Middle Cerebral Artery/complications , Analysis of Variance , Animals , Anti-Inflammatory Agents , Brain Infarction/etiology , Cholera Toxin/pharmacology , Cytokines/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Flow Cytometry , Functional Laterality , Infarction, Middle Cerebral Artery/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Microglia/drug effects , Microglia/pathology , Nervous System Diseases/etiology , RNA, Messenger/metabolism , Rats , Time Factors
10.
J Proteome Res ; 12(7): 3372-82, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23763241

ABSTRACT

To develop a cold chain- and needle/syringe-free rice-based cholera vaccine (MucoRice-CTB) for human use, we previously advanced the MucoRice system by introducing antisense genes specific for endogenous rice storage proteins and produced a molecularly uniform, human-applicable, high-yield MucoRice-CTB devoid of plant-associated sugar. To maintain the cold chain-free property of this vaccine for clinical application, we wanted to use a polished rice powder preparation of MucoRice-CTB without further purification but wondered whether this might cause an unexpected increase in rice allergen protein expression levels in MucoRice-CTB and prompt safety concerns. Therefore, we used two-dimensional fluorescence difference gel electrophoresis and shotgun MS/MS proteomics to compare rice allergen protein expression levels in MucoRice-CTB and wild-type (WT) rice. Both proteomics analyses showed that the only notable change in the expression levels of rice allergen protein in MucoRice-CTB, compared with those in WT rice, was a decrease in the expression levels of α-amylase/trypsin inhibitor-like protein family such as the seed allergen protein RAG2. Real-time PCR analysis showed mRNA of RAG2 reduced in MucoRice-CTB seed. These results demonstrate that no known rice allergens appear to be up-reregulated by genetic modification of MucoRice-CTB, suggesting that MucoRice-CTB has potential as a safe oral cholera vaccine for clinical application.


Subject(s)
Antigens, Plant/genetics , Cholera Toxin/genetics , Cholera/prevention & control , Plant Proteins/genetics , alpha-Amylases/biosynthesis , Administration, Oral , Allergens/genetics , Allergens/isolation & purification , Antigens, Plant/biosynthesis , Cholera/drug therapy , Cholera/pathology , Cholera Toxin/therapeutic use , Cholera Vaccines/administration & dosage , Cholera Vaccines/genetics , Down-Regulation , Gene Expression Regulation, Plant , Humans , Oryza/genetics , Oryza/immunology , Plant Proteins/biosynthesis , Plants, Genetically Modified/genetics , Proteomics , Seeds/genetics , Seeds/metabolism , Tandem Mass Spectrometry , Trypsin Inhibitors/biosynthesis , alpha-Amylases/antagonists & inhibitors
11.
Appl Biochem Biotechnol ; 169(2): 636-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23269637

ABSTRACT

Cholera toxin B-subunit (CTB) has been widely used to facilitate antigen delivery by serving as an effective mucosal carrier molecule for the induction of oral tolerance. However, whether CTB can be used as a transmucosal carrier in the delivery of not only vaccines but also therapeutic proteins has not been widely studied. Thus, we investigate here the concept of receptor-mediated oral delivery of lumbrokinase (LK) proteins which is an important fibrinolytic enzyme derived from earthworm. CTB and LK, separated by a furin cleavage site, was expressed via Pichia pastoris. The activity and proper folding of recombinant protein in yeast were confirmed by Western blot analysis, fibrin plate assays, and G(M1)-ganglioside ELISA. Following oral administration of recombinant protein, the thrombosis model of rats and mice revealed that the oral treatment of rCTB-LK has a more significant anti-thrombotic effect on animals compared with rLK. It is possible to conclude that CTB can successfully enhance its fusion protein LK to be absorbed. The use of CTB as a transmucosal carrier in the delivery of not only vaccines but also therapeutic proteins was supported.


Subject(s)
Cholera Toxin/pharmacokinetics , Endopeptidases/administration & dosage , Endopeptidases/genetics , Mouth Mucosa/metabolism , Pichia/physiology , Thrombosis/prevention & control , Administration, Mucosal , Administration, Oral , Animals , Cholera Toxin/therapeutic use , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacokinetics , Male , Mice , Mice, Inbred BALB C , Protein Engineering/methods , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/administration & dosage , Thrombosis/diagnosis , Treatment Outcome
12.
J Pain ; 11(9): 838-46, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20620120

ABSTRACT

UNLABELLED: Individuals with chronic craniofacial pain experience symptoms that are consistent with central sensitization. In fact, central sensitization may constitute the major disease process in these conditions, particularly if the original injury has healed or the condition is idiopathic. To understand central sensitization we have developed a conjugate of substance P and cholera toxin (SP-CTA). SP-CTA is selectively taken up by cells that express neurokinin receptors. Twenty-four hours following intracisternal administration of SP-CTA, wild-type rats and mice demonstrated signs of persistent background nociception, but when tested for facial cold sensitivity, they did not differ from controls. However, treating the SP-CTA-injected animals with naloxone exposed cold hypersensitivity in the face. Mu-opioid receptor knockout mice treated with SP-CTA demonstrated hypersensitivity without naloxone treatment. These findings suggest that central sensitization leads to activation of an endogenous opioid system. The data also demonstrate that the intracisternal administration of SP-CTA in rodents is a useful model for studying central sensitization as a disease process without having to induce a peripheral injury. PERSPECTIVE: Central sensitization is a concern in many craniofacial pain conditions. In this project, we utilize a conjugate of substance P and the catalytic subunit of cholera toxin to induce central sensitization in the nucleus caudalis of rodents. The data indicate that the injected animals become hypersensitive in the face.


Subject(s)
Cholera Toxin/pharmacology , Substance P/pharmacology , Trigeminal Caudal Nucleus/drug effects , Animals , Behavior, Animal/drug effects , Blotting, Western , Cholera Toxin/therapeutic use , Cisterna Magna , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Facial Pain/drug therapy , Female , Immunohistochemistry , Injections , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement , Rats , Rats, Hairless , Rats, Sprague-Dawley , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/physiology , Substance P/therapeutic use
13.
J Virol ; 84(6): 2983-95, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20053748

ABSTRACT

Prophylactic or therapeutic immunomodulation is an antigen-independent strategy that induces nonspecific immune system activation, thereby enhancing host defense to disease. In this study, we investigated the effect of prophylactic immunomodulation on the outcome of influenza virus infection using three bacterially derived immune-enhancing agents known for promoting distinct immunological profiles. BALB/c mice were treated nasally with either cholera toxin (CT), a mutant form of the CT-related Escherichia coli heat-labile enterotoxin designated LT(R192G), or CpG oligodeoxynucleotide. Mice were subsequently challenged with a lethal dose of influenza A/PR/8/34 virus 24 h after the last immunomodulation treatment and either monitored for survival or sacrificed postchallenge for viral and immunological analysis. Treatment with the three immunomodulators prevented or delayed mortality and weight loss, but only CT and LT(R192G) significantly reduced initial lung viral loads as measured by plaque assay. Analysis performed 4 days postinfection indicated that prophylactic treatments with CT, LT(R192G), or CpG resulted in significantly increased numbers of CD4 T cells, B cells, and dendritic cells and altered costimulatory marker expression in the airways of infected mice, coinciding with reduced expression of pulmonary chemokines and the appearance of inducible bronchus-associated lymphoid tissue-like structures in the lungs. Collectively, these results suggest that, despite different immunomodulatory mechanisms, CT, LT(R192G), and CpG induce an initial inflammatory process and enhance the immune response to primary influenza virus challenge while preventing potentially damaging chemokine expression. These studies provide insight into the immunological parameters and immune modulation strategies that have the potential to enhance the nonspecific host response to influenza virus infection.


Subject(s)
Immunologic Factors/therapeutic use , Influenza, Human , Orthomyxoviridae Infections , Administration, Intranasal , Animals , Bacterial Toxins/administration & dosage , Bacterial Toxins/immunology , Bacterial Toxins/therapeutic use , Body Weight , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Cholera Toxin/therapeutic use , Enterotoxins/administration & dosage , Enterotoxins/immunology , Enterotoxins/therapeutic use , Escherichia coli Proteins/administration & dosage , Escherichia coli Proteins/immunology , Escherichia coli Proteins/therapeutic use , Female , Humans , Immune System/physiology , Immunologic Factors/administration & dosage , Immunologic Factors/immunology , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/drug therapy , Influenza, Human/immunology , Influenza, Human/prevention & control , Lung/cytology , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/therapeutic use , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Survival Rate , Treatment Outcome , Viral Load
14.
J Reprod Immunol ; 81(1): 34-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19501917

ABSTRACT

Chlamydia trachomatis is a significant human pathogen with potentially severe disease sequelae in the genital tract, including infertility. A successful vaccine will need to effectively target immunity to the genital mucosa. Intranasal immunisation with cholera toxin (CT) can target immunity to the genital tract, but has the potential to cause neurological side effects. CTA1-DD is a non-toxic potent mucosal adjuvant which combines the enzymatic properties of CT, with a B cell targeting moiety. Here, we demonstrate that intranasal immunisation with CTA1-DD and chlamydial Major Outer Membrane Protein (MOMP) results in the induction of neutralising systemic and mucosal antibodies, and reduces the level of chlamydial shedding following intravaginal challenge with Chlamydia muridarum. Thus, CTA1-DD is an effective adjuvant for vaccine development against Chlamydia trachomatis, and possibly also a range of other genital pathogens.


Subject(s)
Adjuvants, Immunologic/therapeutic use , B-Lymphocytes/metabolism , Chlamydia Infections/therapy , Chlamydia trachomatis/immunology , Cholera Toxin/therapeutic use , Genitalia/immunology , Mucous Membrane/immunology , Recombinant Fusion Proteins/therapeutic use , Adjuvants, Immunologic/genetics , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Chlamydia Infections/complications , Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Chlamydia muridarum/pathogenicity , Chlamydia trachomatis/pathogenicity , Cholera Toxin/genetics , Cholera Toxin/immunology , Female , Genitalia/microbiology , Genitalia/pathology , Humans , Immunization , Immunoglobulins/blood , Infertility/etiology , Infertility/prevention & control , Mice , Mice, Inbred BALB C , Mucous Membrane/microbiology , Mucous Membrane/pathology , Porins/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
15.
Ann Allergy Asthma Immunol ; 102(4): 314-22, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19441603

ABSTRACT

BACKGROUND: Recent studies have shown that by acting as strong TH1 response-inducing adjuvants, DNA immunostimulatory sequence oligdeoxynucleotides (ISS-ODNs) can be used in the treatment of allergic diseases, and efforts are being made to enhance these TH1 adjuvant actions. OBJECTIVE: To determine whether intranasally delivered ISS-ODN/cholera toxin B (CTB) conjugate has enhanced antiallergic effects in an allergic rhinitis mouse model. METHODS: BALB-c mice were sensitized with ovalbumin. Chemical conjugation of ISS-ODN and CTB was performed. After a single local intranasal administration of 50 microg of ISS-ODN or high- and low-dose (50- and 5-microg) ISS-ODN/CTB conjugate, we measured the allergic response in terms of sneezing events, eosinophil infiltration in the nasal mucosa, serum ovalbumin specific IgE and IgG2a levels, and TH1 and TH2 cytokine levels in nasal lavage fluid and spleen cell cultures. RESULTS: A single local administration of 50 microg of ISS-ODN did not suppress the allergic phenotype. However, 50 and 5 microg of ISS-ODN/CTB conjugate significantly attenuated allergic symptoms, eosinophil infiltration in the nasal mucosa, and interleukin 4 production from nasal lavage fluid and cultured splenocyte supernatant compared with the allergic control. Serum specific IgG2a and interleukin 12 production in nasal lavage fluid and spleen cell cultures was significantly increased. CONCLUSIONS: In a mouse model of allergic rhinitis, a single intranasal delivery of low-dose ISS-ODN/CTB conjugate effectively protects previously sensitized mice from allergic hypersensitivity responses. With further research, ISS-ODN/CTB conjugate may serve as a new allergen-independent intranasal vaccine for the treatment of allergic rhinitis.


Subject(s)
Cholera Toxin/therapeutic use , Immunotoxins/therapeutic use , Oligodeoxyribonucleotides/therapeutic use , Rhinitis, Allergic, Perennial/therapy , Adjuvants, Immunologic/therapeutic use , Administration, Intranasal , Animals , Cholera Toxin/immunology , DNA/immunology , DNA/therapeutic use , Eosinophils/immunology , Eosinophils/pathology , Female , Immunoglobulin G/blood , Immunotoxins/immunology , Interleukin-12/analysis , Interleukin-4/analysis , Mice , Mice, Inbred BALB C , Nasal Lavage Fluid/immunology , Nasal Mucosa/immunology , Nasal Mucosa/pathology , Oligodeoxyribonucleotides/immunology , Ovalbumin/blood , Ovalbumin/immunology , Rhinitis, Allergic, Perennial/immunology , Rhinitis, Allergic, Perennial/pathology , Vaccines, Conjugate/immunology , Vaccines, Conjugate/therapeutic use
16.
Arthritis Rheum ; 60(6): 1672-82, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19479868

ABSTRACT

OBJECTIVE: To determine whether a cholera toxin-derived, novel immunomodulating fusion protein, CTA1R7K-COL-DD, carrying the class II major histocompatibility complex H-2q-restricted type II collagen peptide aa 259-274, can induce therapeutic tolerance and prevent collagen-induced arthritis (CIA) when administered intranasally in DBA/1 mice, and to assess whether ADP-ribosylation at the mucosal membranes exerts a regulatory function such that the outcome of tolerance or immune enhancement can be controlled. METHODS: DBA/1 mice with CIA were treated intranasally with CTA1R7K-COL-DD. The therapeutic effect was monitored for 46 days after the onset of disease. Clinical scoring of disease, histologic examination of inflammation, and bone erosion were assessed, and cytokine levels were determined in the serum or supernatants from splenocytes stimulated with recall antigen. RESULTS: The protective effect of CTA1R7K-COL-DD resulted in roughly 60% of the mice having no clinical signs or histologic evidence of disease after treatment, and those with CIA had significantly milder disease with less bone erosion. The protective status was associated with lower serum titers of IgG1, IgG2a, IgG2b, and IgG3 anticollagen and a substantial decrease in the production of interleukin-6 (IL-6), IL-17, and interferon-gamma, while levels of IL-10 were markedly up-regulated both in the serum and at the T cell level. CONCLUSION: The enzymatically inactive mutant fusion protein CTA1R7K-COL-DD provided substantial therapeutic protection against CIA following intranasal administration. The mechanism behind the effect appears to be mediated by peptide-specific regulatory T cells induced by mucosal exposure to the peptide containing CTA1R7K-COL-DD vector. In addition, ADP-ribosylation at the mucosal membranes acts as a key regulator controlling mucosal tolerance or immunity.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Experimental/prevention & control , Cholera Toxin/therapeutic use , Drug Tolerance/physiology , Mucous Membrane/metabolism , Recombinant Fusion Proteins/therapeutic use , ADP Ribose Transferases/metabolism , Administration, Intranasal , Animals , Arthritis, Experimental/chemically induced , CD4-Positive T-Lymphocytes/metabolism , Cholera Toxin/administration & dosage , Cholera Toxin/genetics , Disease Models, Animal , Immunoglobulin G/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred DBA , Peptide Fragments/genetics , Peptide Fragments/physiology , Peptide Fragments/therapeutic use , Plasmids , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics
17.
Mucosal Immunol ; 2(4): 331-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19404246

ABSTRACT

In healthy individuals, humoral immune responses to allergens consist of serum IgA and IgG4, whereas cellular immune responses are controlled by regulatory T (Treg) cells. In search of new compounds that might prevent the onset of allergies by stimulating this type of immune response, we have focused on the mucosal adjuvant, cholera toxin B (CTB), as it induces the formation of Treg cells and production of IgA. Here, we have found that CTB suppresses the potential of dendritic cells to prime for Th2 responses to inhaled allergen. When we administered CTB to the airways of naïve and allergic mice, it strongly suppressed the salient features of asthma, such as airway eosinophilia, Th2 cytokine synthesis, and bronchial hyperreactivity. This beneficial effect was only transferable to other mice by transfer of B but not of T lymphocytes. CTB caused a transforming growth factor-beta-dependent rise in antigen-specific IgA in the airway luminal secretions, which was necessary for its preventive and curative effect, as all effects of CTB were abrogated in mice lacking the luminal IgA transporting polymeric Ig receptor. Not only do these findings show a novel therapeutic avenue for allergy, they also help to explain the complex relationship between IgA levels and risk of developing allergy in humans.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cholera Toxin/therapeutic use , Dendritic Cells/immunology , Hypersensitivity/therapy , Immunoglobulin A, Secretory/immunology , Adoptive Transfer , Allergens/immunology , Animals , B-Lymphocytes/immunology , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Female , Hypersensitivity/immunology , Immunoglobulin A, Secretory/biosynthesis , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/immunology
18.
J Immunol ; 182(5): 2610-9, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234155

ABSTRACT

Solitary intestinal lymphoid tissue (SILT) comprises a spectrum of phenotypically diverse lymphoid aggregates interspersed throughout the small intestinal mucosa. Manifestations of SILT range from tiny lymphoid aggregates almost void of mature lymphocytes to large structures dominated by B cells. Large SILT phenotypically resemble a single Peyer's patch follicle, suggesting that SILT might contribute to intestinal humoral immune responses. In this study, we track the fate of individual SILT in vivo over time and analyze SILT formation and function in chemokine receptor CXCR5-deficient mice. We show that, in analogy to Peyer's patches, formation of SILT is invariantly determined during ontogeny and depends on CXCR5. Young CXCR5-deficient mice completely lack SILT, suggesting that CXCR5 is essential for SILT formation during regular postnatal development. However, microbiota and other external stimuli can induce the formation of aberrant SILT distinguished by impaired development of B cell follicles in CXCR5-deficient mice. Small intestinal transplantation and bone marrow transplantation reveal that defect follicle formation is due to impaired B cell homing. Moreover, oral immunization with cholera toxin or infection with noninvasive Salmonella fail to induce efficient humoral immune responses in CXCR5-deficient mice. Bone marrow transplantation of CXCR5-deficient recipients with wild-type bone marrow rescued B cell follicle formation in SILT but failed to restore full humoral immune responses. These results reveal an essential role of CXCR5 in Peyer's patch and SILT development and function and indicate that SILT do not fully compensate for the lack of Peyer's patches in T cell-dependent humoral immune responses.


Subject(s)
B-Lymphocytes/immunology , Chemotaxis, Leukocyte/immunology , Immunoglobulin A/biosynthesis , Intestinal Mucosa/immunology , Lymphoid Tissue/immunology , Receptors, CXCR5/physiology , Administration, Oral , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Bone Marrow Transplantation/immunology , Cholera Toxin/therapeutic use , Intestinal Mucosa/metabolism , Lymphoid Tissue/growth & development , Lymphoid Tissue/metabolism , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/immunology , T-Lymphocytes/immunology
19.
Am J Pathol ; 173(5): 1243-52, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18832585

ABSTRACT

The anatomical and functional arrangement of the gastrointestinal tract suggests that this organ, beside its digestive and absorptive functions, regulates the trafficking of macromolecules between the environment and the host through a barrier mechanism. Under physiological circumstances, this trafficking is safeguarded by the competency of intercellular tight junctions, structures whose physiological modulation is mediated by, among others, the recently described protein zonulin. To prevent harm and minimize inflammation, the same paracellular pathway, in concert with the gut-associated lymphoid tissue and the neuroendocrine network, controls the equilibrium between tolerance and immunity to nonself antigens. The zonulin pathway has been exploited to deliver drugs, macromolecules, or vaccines that normally would not be absorbed through the gastrointestinal mucosal barrier. However, if the tightly regulated trafficking of macromolecules is jeopardized secondary to prolonged zonulin up-regulation, the excessive flow of nonself antigens in the intestinal submucosa can cause both intestinal and extraintestinal autoimmune disorders in genetically susceptible individuals. This new paradigm subverts traditional theories underlying the development of autoimmunity, which are based on molecular mimicry and/or the bystander effect, and suggests that the autoimmune process can be arrested if the interplay between genes and environmental triggers is prevented by re-establishing intestinal barrier competency. Understanding the role of zonulin-dependent intestinal barrier dysfunction in the pathogenesis of autoimmune diseases is an area of translational research that encompasses many fields.


Subject(s)
Cholera Toxin/therapeutic use , Intestines/pathology , Intestines/physiology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Cholera Toxin/pharmacology , Haptoglobins , Humans , Immunity, Mucosal/drug effects , Intestinal Mucosa/drug effects , Permeability/drug effects , Protein Precursors , Tight Junctions/metabolism
20.
Mol Biotechnol ; 32(1): 1-15, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16382177

ABSTRACT

Several bacterial and plant enterotoxin B subunit-islet autoantigen fusion proteins were compared for their ability to serve as islet autoantigen carriers and adjuvants for reduction of pancreatic islet inflammation associated with type 1 diabetes. The cholera toxin B subunit (CTB), the heat-labile toxin B subunit from enterotoxigenic Escherichia coli (LTB), the Shigella toxin B subunit (STB), and the plant toxin ricin B subunit (RTB) were genetically linked to the islet autoantigens proinsulin (INS) and glutamic acid decarboxylase (GAD). The adjuvant-autoantigen gene fusions were transferred to a bacterial expression vector and the corresponding fusion proteins synthesized in E. coli. The purified adjuvant-autoantigen proteins were fed to 5-wk-old nonobese diabetic (NOD) mice once a week for 4 wk. Histological examination of pancreatic islets isolated from inoculated mice showed significant levels of insulitis reduction in comparison with uninoculated mice. The ratio of serum anti-INS and anti-GAD IgG2c to IgG1 antibody isotype titers increased in all ligand-autoantigen inoculated animal groups, suggesting an increase in effector Th2 lymphocytes in B subunit-mediated insulitis suppression. The results of these experiments indicate that bacterial and plant enterotoxin B subunit ligand-autoantigens enhance insulitis reduction in NOD mice. This research prompts further exploration of a multiadjuvant/autoantigen co-delivery strategy that may facilitate type 1 diabetes prevention and suppression in animals and humans.


Subject(s)
Autoantigens/therapeutic use , Diabetes Mellitus, Type 1/therapy , Enterotoxins/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Animals , Antibody Formation/immunology , Asialoglycoproteins/metabolism , Autoantigens/genetics , Autoantigens/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/therapeutic use , Cholera Toxin/genetics , Cholera Toxin/immunology , Cholera Toxin/therapeutic use , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Enterotoxins/genetics , Enterotoxins/immunology , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Escherichia coli Proteins/therapeutic use , Female , Fetuins , G(M1) Ganglioside/metabolism , Glutamate Decarboxylase/genetics , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin Isotypes/blood , Immunoglobulin Isotypes/immunology , Immunotherapy, Active/methods , Islets of Langerhans/chemistry , Mice , Mice, Inbred NOD , Peptide Fragments/genetics , Proinsulin/genetics , Protein Subunits/genetics , Protein Subunits/immunology , Protein Subunits/therapeutic use , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Ricin/genetics , Ricin/immunology , Shiga Toxin/genetics , Shiga Toxin/immunology , Shiga Toxin/therapeutic use , Trihexosylceramides/metabolism , alpha-Fetoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...