Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
J Nanobiotechnology ; 16(1): 59, 2018 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-30077180

RESUMEN

BACKGROUND: Various nanocarriers have been used to deliver subunit vaccines specifically to dendritic cells (DCs) for the improvement of immunogenicity. However, due to their insufficient DC priming ability, these vaccines could not elicit effective innate immunity. We have recently developed a DC-targeting bio-nanocapsule (BNC) by displaying anti-CD11c IgGs via protein A-derived IgG Fc-binding Z domain on the hepatitis B virus envelope L protein particles (α-DC-ZZ-BNC). RESULTS: After the chemical modification with antigens (Ags), the α-DC-ZZ-BNC-Ag complex could deliver Ags to DCs efficiently, leading to effective DC maturation and efficient endosomal escape of Ags, followed by Ag-specific T cell responses and IgG productions. Moreover, the α-DC-ZZ-BNC modified with Japanese encephalitis virus (JEV) envelope-derived D3 Ags could confer protection against 50-fold lethal dose of JEV injection on mice. CONCLUSION: The α-DC-ZZ-BNC-Ag platform was shown to induce humoral and cellular immunities effectively without any adjuvant.


Asunto(s)
Antígeno CD11c/inmunología , Células Dendríticas/inmunología , Inmunogenicidad Vacunal , Vacunas contra la Encefalitis Japonesa/inmunología , Nanocápsulas/química , Animales , Antígenos Virales/administración & dosificación , Antígenos Virales/inmunología , Línea Celular , Células Dendríticas/metabolismo , Virus de la Encefalitis Japonesa (Especie)/química , Virus de la Encefalitis Japonesa (Especie)/fisiología , Humanos , Inmunidad Celular , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Vacunas contra la Encefalitis Japonesa/administración & dosificación , Ratones Endogámicos BALB C , Ovalbúmina/química , Tamaño de la Partícula , Proteína Estafilocócica A/química , Proteínas del Envoltorio Viral/química
2.
Microbiol Immunol ; 59(12): 735-43, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26577130

RESUMEN

Although the BCG vaccine can prevent tuberculosis (TB) in infants, its ability to prevent adult pulmonary TB is reportedly limited. Therefore, development of a novel effective vaccine against pulmonary TB has become an international research priority. We have previously reported that intranasal vaccination of mice with a mycobacterial heparin-binding hemagglutinin adhesin (HBHA) plus mucosal adjuvant cholera toxin (CT) enhances production of IFN-γ and anti-HBHA antibody and suppresses extrapulmonary bacterial dissemination after intranasal infection with BCG. In the present study, the effects of intranasal HBHA + CT vaccine on murine pulmonary Mycobacterium tuberculosis (Mtb) infection were examined. Intranasal HBHA + CT vaccination alone failed to reduce the bacterial burden in the infected lung. However, a combination vaccine consisting of s.c. BCG priming and an intranasal HBHA + CT booster significantly enhanced protective immunity against pulmonary Mtb infection on day 14 compared with BCG vaccine alone. Further, it was found that intranasal HBHA + CT vaccine enhanced not only IFN-γ but also IL-17A production by HBHA-specific T cells in the lung after pulmonary Mtb infection. Therefore, this combination vaccine may be a good candidate for a new vaccine strategy against pulmonary TB.


Asunto(s)
Adhesinas Bacterianas/inmunología , Vacuna BCG/farmacología , Lectinas/inmunología , Pulmón/inmunología , Mycobacterium tuberculosis/inmunología , Células Th17/inmunología , Tuberculosis Pulmonar/prevención & control , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos Bacterianos/inmunología , Vacuna BCG/inmunología , Toxina del Cólera/inmunología , Femenino , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Lectinas/genética , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Linfocitos T/metabolismo , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología
3.
J Pediatr Hematol Oncol ; 37(6): e375-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25929611

RESUMEN

Hemolytic disease of the newborn (HDN) arising from MNSs incompatibility is rare, with few reports of prolonged anemia and reticulocytopenia following HDN. We report the younger of 2 male siblings, both of whom had anti-M-induced HDN and anemia persisting for over a month. Peripheral reticulocytes remained inappropriately low for the degree of anemia, and they needed multiple red cell transfusions. Viral infections were ruled out. Corticosteroids were given for suspected pure red cell aplasia. Anemia and reticulocytopenia subsequently improved. Colony-forming unit erythroid assay revealed erythropoietic suppression of M antigen-positive erythroid precursor cells cultured with maternal or infant sera containing anti-M. In conclusion, maternal anti-M caused HDN and prolonged anemia by erythropoietic suppression in 2 siblings.


Asunto(s)
Anemia/etiología , Eritroblastosis Fetal/etiología , Células Precursoras Eritroides/patología , Eritropoyesis/inmunología , Inmunoglobulina M/inmunología , Isoanticuerpos/inmunología , Aplasia Pura de Células Rojas/complicaciones , Adulto , Anemia/patología , Diferenciación Celular , Ensayo de Unidades Formadoras de Colonias , Eritroblastosis Fetal/patología , Células Precursoras Eritroides/inmunología , Femenino , Humanos , Inmunoglobulina M/sangre , Lactante , Recién Nacido , Masculino , Embarazo , Pronóstico , Hermanos
4.
BMC Biotechnol ; 14: 38, 2014 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-24884459

RESUMEN

BACKGROUND: Lactobacillus species are used as bacterial vectors to deliver functional peptides to the intestine because they are delivered live to the intestine, colonize the mucosal surface, and continue to produce the desired protein. Previously, we generated a recombinant Lactobacillus casei secreting the cholera toxin B subunit (CTB), which can translocate into intestinal epithelial cells (IECs) through GM1 ganglioside. Recombinant fusion proteins of CTB with functional peptides have been used as carriers for the delivery of these peptides to IECs because of the high cell permeation capacity of recombinant CTB (rCTB). However, there have been no reports of rCTB fused with peptides expressed or secreted by Lactobacillus species. In this study, we constructed L. casei secreting a recombinant fusion protein of CTB with YVAD (rCTB-YVAD). YVAD is a tetrapeptide (tyrosine-valine-alanine-aspartic acid) that specifically inhibits caspase-1, which catalyzes the production of interleukin (IL)-1ß, an inflammatory cytokine, from its inactive precursor. Here, we examined whether rCTB-YVAD secreted by L. casei binds to GM1 ganglioside and inhibits caspase-1 activation in Caco-2 cells used as a model of IECs. RESULTS: We constructed the rCTB-YVAD secretion vector pSCTB-YVAD by modifying the rCTB secretion vector pSCTB. L. casei secreting rCTB-YVAD was generated by transformation with pSCTB-YVAD. Both the culture supernatant of pSCTB-YVAD-transformed L. casei and purified rCTB-YVAD bound to GM1 ganglioside, as did the culture supernatant of pSCTB-transformed L. casei and purified rCTB. Interestingly, although both purified rCTB-YVAD and rCTB translocated into Caco-2 cells, regardless of lipopolysaccharide (LPS), only purified rCTB-YVAD but not rCTB inhibited LPS-induced caspase-1 activation and subsequent IL-1ß secretion in Caco-2 cells, without affecting cell viability. CONCLUSIONS: The rCTB protein fused to a functional peptide secreted by L. casei can bind to GM1 ganglioside, like rCTB, and recombinant YVAD secreted by L. casei may exert anti-inflammatory effects in the intestine. Therefore, rCTB secreted by L. casei has potential utility as a vector for the delivery of YVAD to IECs.


Asunto(s)
Caspasa 1/metabolismo , Toxina del Cólera/metabolismo , Interleucina-1beta/metabolismo , Lacticaseibacillus casei/metabolismo , Oligopéptidos/metabolismo , Secuencia de Aminoácidos , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Toxina del Cólera/genética , Toxina del Cólera/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Lipopolisacáridos/toxicidad , Oligopéptidos/química , Oligopéptidos/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología
5.
Vaccine ; 42(7): 1757-1767, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38365487

RESUMEN

Vaccines against Shiga toxin (Stx)-producing Escherichia coli (STEC) have not yet been developed. Two immunologically distinct serotypes of Stx (Stx1 and Stx2) are the main virulence factors of STEC. Thus, blocking their B subunits (StxB) from binding to the cell surface receptor globotriaosylceramide (Gb3) efficiently prevents the action of these toxins. We expressed Stx1B and Stx2B in E. coli inclusion bodies and reassembled them into pentamers by a stepwise dialysis. Stx1B pentamer fully protected mice against Stx1 challenge, but Stx2B pentamer failed to protect mice against Stx2 challenge. To explain those observations, we proposed that the pentamer of Stx2B readily dissociates into its constituent monomers, especially under in vivo conditions, thus being unable to induce pentamer-specific immunity. To increase pentamer stability, we fused the B subunit to a pentameric coiled-coil domain of the cartilage oligomeric matrix protein (COMP). This "five-to-five" fusion hybrid molecule (Stx2B-COMP) was shown to be protective against Stx2 challenge, demonstrating that the Stx2B subunit when leashed and bundled by a rigid pentameric coiled-coil domain mount a pentamer-specific immune response and efficiently neutralize the toxin both in vitro and in vivo. Our data strongly suggest that the Stx2B subunit moiety fluctuates between a pentameric and monomeric state within the fusion protein, which may increase the likelihood of the immune system recognizing the pentameric conformation for toxin neutralization.


Asunto(s)
Infecciones por Escherichia coli , Vacunas , Ratones , Animales , Escherichia coli , Infecciones por Escherichia coli/prevención & control
6.
Microbiol Immunol ; 57(6): 470-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23773026

RESUMEN

An engineered bio-nanocapsule (BNC) comprising modified hepatitis B surface antigen L protein was used as a physical scaffold for envelope protein domain III (D3) of Japanese encephalitis virus (JEV). At the N terminus, the BNC contained a two-tandem repeat of the Z domain (ZZ) derived from Staphylococcus aureus protein A (ZZ-BNC). The Lys-rich ZZ moiety exposed on the surface of ZZ-BNC was used for chemical conjugation with the JEV D3 antigen, which had been expressed and purified from Escherichia coli. Immunization of mice with D3 loaded on the surface of ZZ-BNC (ZZ-BNC:D3) augmented serum IgG response against JEV and increased protection against lethal JEV infection. The present study suggests that innocuous recombinant antigens, when loaded on the surface of ZZ-BNC, can be transformed to immunogenic antigens.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Virus de la Encefalitis Japonesa (Especie)/inmunología , Vacunas contra la Encefalitis Japonesa/inmunología , Nanocápsulas/administración & dosificación , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/sangre , Virus de la Encefalitis Japonesa (Especie)/genética , Escherichia coli/genética , Femenino , Inmunoglobulina G/sangre , Vacunas contra la Encefalitis Japonesa/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/metabolismo , Análisis de Supervivencia , Vacunación/métodos , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética
7.
Biol Pharm Bull ; 36(6): 952-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23727916

RESUMEN

Lactic acid bacteria (LAB) are used in various fields, including in food and medical supplies. There has been a great deal of research into vaccine development using LAB as carriers due to their "generally recognized as safe" status. Cholera is an infectious disease that causes diarrhea due to cholera toxin (CT) produced by Vibrio cholerae. The pentameric cholera toxin B (CTB) subunit has no toxicity, and is used as an antigen in cholera vaccines and as a delivery molecule in vaccines to various diseases. In this study, we generated recombinant LAB expressing and secreting CTB. Here, we first report that CTB expressed and secreted from LAB bound to GM1 ganglioside. The secreted CTB was purified, and its immunogenicity was determined by intranasal administration into mice. The results of the present study suggested that it may be useful as the basis of a new oral cholera vaccine combining LAB and CTB.


Asunto(s)
Antígenos Bacterianos/metabolismo , Toxina del Cólera/metabolismo , Lactobacillus/metabolismo , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Toxina del Cólera/genética , Toxina del Cólera/inmunología , Vacunas contra el Cólera/administración & dosificación , Escherichia coli/genética , Femenino , Gangliósidos/metabolismo , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos BALB C , Plásmidos , Proteínas Recombinantes/metabolismo
8.
Clin Pediatr Endocrinol ; 31(1): 44-49, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35002068

RESUMEN

X-linked nephrogenic diabetes insipidus (NDI) is caused by variations in arginine vasopressin receptor 2 (AVPR2). Some patients show partial resistance to arginine vasopressin (AVP). A 19-month-old Japanese boy presented with polydipsia since infancy. His mother had a history of polydipsia during pregnancy, and his maternal granduncle also had polydipsia. Intermediate urine osmolality and markedly high plasma AVP levels were observed in the water deprivation test. Subsequent pitressin administration caused no further elevation in urine osmolality. We diagnosed the patient with partial NDI, initiated therapy with hydrochlorothiazide, and placed him on a low-sodium diet. Although his urine volume decreased by 20-30% after the initiation of therapy, progressive hydronephrosis and growth retardation developed 2 years later. We investigated his genetic background by multiplex targeted sequencing of genes associated with inherited renal diseases, including AVPR2 and aquaporin-2 (AQP2). We identified a hemizygous missense variant in AVPR2 NM_000054:c.371A>G,p.(Tyr124Cys) in the boy and a heterozygous variant in the mother at the same locus. Distinguishing partial NDI from primary polydipsia is difficult because of its mild symptoms. Markedly elevated plasma AVP levels with intermediate urine osmolality may suggest partial NDI, and genetic analysis can be useful for such patients.

9.
Infect Immun ; 79(10): 4260-75, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21807905

RESUMEN

The creation of subunit vaccines to prevent malaria infection has been hampered by the intrinsically weak immunogenicity of the recombinant antigens. We have developed a novel strategy to increase immune responses by creating genetic fusion proteins to target specific antigen-presenting cells (APCs). The fusion complex was composed of three physically linked molecular entities: (i) a vaccine antigen, (ii) a multimeric α-helical coiled-coil core, and (iii) an APC-targeting ligand linked to the core via a flexible linker. The vaccine efficacy of the tricomponent complex was evaluated using an ookinete surface protein of Plasmodium vivax, Pvs25, and merozoite surface protein-1 of Plasmodium yoelii. Immunization of mice with the tricomponent complex induced a robust antibody response and conferred substantial levels of P. vivax transmission blockade as evaluated by a membrane feed assay, as well as protection from lethal P. yoelii infection. The observed effect was strongly dependent on the presence of all three components physically integrated as a fusion complex. This system, designated the tricomponent immunopotentiating system (TIPS), onto which any recombinant protein antigens or nonproteinaceous substances could be loaded, may be a promising strategy for devising subunit vaccines or adjuvants against various infectious diseases, including malaria.


Asunto(s)
Diseño de Fármacos , Vacunas contra la Malaria/administración & dosificación , Malaria/prevención & control , Plasmodium vivax/inmunología , Plasmodium yoelii/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas Sintéticas/administración & dosificación , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Antígenos de Superficie/química , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Linfocitos B/inmunología , Secuencia de Bases , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/inmunología , Femenino , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/inmunología , Ligandos , Activación de Linfocitos , Malaria/inmunología , Vacunas contra la Malaria/química , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Malaria Vivax/inmunología , Malaria Vivax/prevención & control , Proteínas Matrilinas , Proteína 1 de Superficie de Merozoito/química , Proteína 1 de Superficie de Merozoito/genética , Proteína 1 de Superficie de Merozoito/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Plasmodium vivax/genética , Plasmodium yoelii/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Vacunas de Subunidad/química , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/química , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
10.
Int Immunol ; 22(4): 307-18, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20167585

RESUMEN

Although the importance of T(h)1-type immune response in protection against mycobacterial infection is well recognized, its regulatory mechanism in the Mycobacterium tuberculosis (Mtb)-infected lung is not well characterized. To address this issue, we analyzed kinetics of induction of mycobacterial antigen-specific CD4(+) T(h)1 T cells after mycobacterial infection in P25 TCR-transgenic (Tg) mice which express TCR alpha and beta chains from a mycobacterial Ag85B-specific MHC class II A(b)-restricted CD4(+) T-cell clone. To supply normal regulatory T-cell repertoire, we transferred normal spleen T cells into the P25 TCR-Tg mice before infection. High dose subcutaneous infection with Mtb or Mycobacterium bovis bacillus Calmette-Guérin (BCG) induced P25 TCR-Tg CD4(+) T(h)1 cells within a week. In contrast, high-dose Mtb or BCG infection into the lung failed to induce P25 TCR-Tg CD4(+) T(h)1 cells at the early stage of the infection. Furthermore, low-dose Mtb infection into the lung induced P25 TCR-Tg CD4(+) T(h)1 cells on day 21 in the mediastinal lymph node but not in the lung. IL-10 was partially involved in the suppression of T(h)1 induction in the lung because pretreatment of mice with anti-IL-10 antibody resulted in increase of P25 TCR-Tg CD4(+) T(h)1 cells in the Mtb-infected lung on day 21 of the infection, whereas neutralization of transforming growth factor-beta, another important suppressive cytokine in the lung, showed no effects on the T(h)1 induction. Our data suggest that induction of anti-mycobacterial CD4(+) T(h)1 cells is suppressed in the mycobacteria-infected lung partially by IL-10.


Asunto(s)
Antígenos Bacterianos/inmunología , Linfocitos T CD4-Positivos/inmunología , Pulmón/inmunología , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Regulación hacia Abajo , Interleucina-10/inmunología , Pulmón/microbiología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Especificidad del Receptor de Antígeno de Linfocitos T
11.
Brain Dev ; 43(7): 804-808, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33827760

RESUMEN

INTRODUCTION: SCN8A-related epilepsy has various phenotypes. In particular, patients with developmental and epileptic encephalopathy (DEE) are resistant to antiepileptic drugs and may present with autonomic symptoms, such as marked bradycardia and apnea during seizures, and thus have an increased risk of sudden death. Herein, we report a case of very severe SCN8A-related epilepsy necessitating cardiac pacemaker implantation because of repetitive ictal asystole. CASE REPORT: The patient was a 14-month-old girl. Tremor and generalized tonic seizure occurred after birth. During seizures, bradycardia and perioral cyanosis occurred, and then, after developing tachycardia and apnea, marked bradycardia and generalized cyanosis occurred, which sometimes resulted in ictal asystole requiring cardiopulmonary resuscitation. Her seizures were refractory to antiepileptic drugs. As the seizures requiring resuscitation did not decrease, cardiac pacemaker implantation was performed four months after birth. Exome sequencing revealed a heterozygous de novo variant in SCN8A (NM_014191.3:c.4934T>C,p.(Met1645Thr)). Even though phenytoin was effective, seizures with bradycardia remained approximately once a month, and pacemaker activity was observed. CONCLUSIONS: This is, to our knowledge, the first reported case of SCN8A-related DEE in whom pacemaker implantation was performed. Pacemaker implantation should be considered as a treatment option for critical patients with SCN8A-related DEE as in the present case, because the incidence of sudden unexpected death in epilepsy is reported to be approximately 10% in patients with SCN8A-related DEE.


Asunto(s)
Encefalopatías/genética , Epilepsia/genética , Paro Cardíaco/terapia , Canal de Sodio Activado por Voltaje NAV1.6/genética , Trastornos del Neurodesarrollo/genética , Marcapaso Artificial , Epilepsia/complicaciones , Femenino , Paro Cardíaco/etiología , Humanos , Lactante
12.
Infect Immun ; 78(9): 3773-82, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20584978

RESUMEN

The nontoxic cholera toxin B subunit (CTB) was evaluated as a potential delivery molecule for the Plasmodium vivax ookinete surface protein, Pvs25. Recombinant Pvs25 was expressed as a secreted protein in the yeast Pichia pastoris, as a mixture of isoforms including multimers and the A and B monomers. The A isoform with the presumed native protein fold was the most abundant, accounting for more than 40% of all expressed protein. The molecularly uniform A isoform was chemically conjugated to CTB via its primary amines, and the fusion protein, retaining GM1-ganglioside affinity, was administered to BALB/c mice by the subcutaneous (s.c.) or intranasal (i.n.) route. Immunization of mice with conjugated Pvs25 without supplemental adjuvant induced antisera that specifically recognized P. vivax ookinetes in vitro. Furthermore, the antisera, when mixed with parasitized blood isolated from P. vivax patients from Thailand, was found to reduce parasite transmission to mosquitoes, conferring a 93 to 98% (s.c.) or a 73 to 88% (i.n.) decrease in oocyst number. Unconjugated Pvs25 alone conferred only a 23 to 60% (s.c.) or a 0 to 6% (i.n.) decrease in oocyst number. Coadministration of extraneous adjuvants, however, further enhanced the vaccine efficacy up to complete blockade. Taken together, we conclude that a weakly immunogenic Pvs25 by itself, when linked to CTB, transforms into a potent transmission-blocking antigen in both i.n. and s.c. routes. In addition, the present study is, to the best of our knowledge, the first demonstration of the immune potentiating function of CTB for a vaccine antigen delivered by the s.c. route.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos de Protozoos/inmunología , Antígenos de Superficie/inmunología , Toxina del Cólera/administración & dosificación , Vacunas contra la Malaria/inmunología , Malaria Vivax/transmisión , Plasmodium vivax/inmunología , Administración Intranasal , Animales , Anopheles , Femenino , Humanos , Inyecciones Subcutáneas , Vacunas contra la Malaria/administración & dosificación , Malaria Vivax/prevención & control , Ratones , Ratones Endogámicos BALB C , Vacunas Sintéticas/inmunología
13.
Infect Immun ; 77(12): 5496-500, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19752035

RESUMEN

Malaria vaccines based on ookinete surface proteins (OSPs) of the malaria parasites block oocyst development in feeding mosquitoes and hence disrupt the parasite life cycle and prevent the disease from being transmitted to other individuals. To investigate whether a noninvasive mucosal vaccination regimen effectively blocks parasite transmission in vivo, Plasmodium yoelii Pys25, a homolog of the Pfs25 and Pvs25 OSPs of Plasmodium falciparum and Plasmodium vivax, respectively, was intranasally (i.n.) administered using a complement-deficient DBA/2 mouse malaria infection model, in which a highly elevated level of oocysts develops in feeding mosquitoes. Vaccinated mice developed a robust antibody response when the vaccine antigen was given together with cholera toxin adjuvant. The induced immune serum was passively transferred to DBA/2 mice 3 days after infection with P. yoelii 17XL, and Anopheles stephensi mosquitoes were allowed to feed on the infected mice before or after serum transfusion. This passive immunization completely blocked oocyst development; however, immune serum induced by the antigen or adjuvant alone did not have such a profound antiparasite effect. Further, when i.n. vaccinated mice were infected with the parasite and then mosquitoes were allowed to directly feed on the infected mice, complete blockage of transmission was again observed. To our knowledge, this is the first time that mucosal vaccination has been demonstrated to be efficacious for directly preventing parasite transmission from vaccinated animals to mosquitoes, and the results may provide important insight into rational design of nonparenteral vaccines for use against human malaria.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Malaria/transmisión , Proteínas Protozoarias/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Anopheles/fisiología , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/administración & dosificación , Toxina del Cólera/administración & dosificación , Vectores de Enfermedades , Femenino , Vacunas contra la Malaria/administración & dosificación , Ratones , Ratones Endogámicos DBA , Plasmodium yoelii/inmunología , Proteínas Protozoarias/administración & dosificación
14.
Parasitol Int ; 58(1): 36-44, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18940265

RESUMEN

Experimental vaccination with radiation-attenuated cercariae (RAC) confers possible practical levels of resistance to challenge infection by humoral and by cellular mechanism. Here, we aimed to identify possible vaccine antigens by using specific IgG antibody from RAC vaccinated miniature pig. Two milligrams of soluble egg antigen (SEA) or schistosomal worm antigen preparation (SWAP) was fractionated using two dimensional liquid chromatography (proteome PF 2D) consisted of high performance chromatofocusing (HPCF) and high resolution reversed phase chromatography (HPRP). Of the 42 HPCF fractions of SEA or SWAP, 26 (61.9%) or 15 (35.7%) showed positive dot blot reaction with RAC vaccinated serum respectively. The dot blot positive fractions were applied to the second HPRP column. One hundred and seven out of 26 x 96 of SEA fractions and 18 out of 15 x 96 SWAP fractions reacted with RAC vaccinated serum. From the positive fractions we chose 17 of SEA and 10 of SWAP that had no reactivity with normal cercariae infected (NCI) sera and had single peak of 214 nm; and automated N-terminal amino acid sequence based on in situ Edman Reaction was conducted. Four sequences were obtained and applied to the homology search in NCBI database. A total of eight candidate genes were listed up and their cDNA clones from schistosomula stage were obtained. Two of the recombinant proteins (AAW27472.1 and AXX25883.1) showed strong reactivity with the RAC vaccinated serum but marginal with NCI serum. This protocol using proteome PF 2D could be applicable in identifying immunoreactive proteins from crude extract for the development of vaccines or for diagnostics.


Asunto(s)
Anticuerpos Antihelmínticos/inmunología , Antígenos Helmínticos/inmunología , Proteínas del Helminto , Proteoma , Schistosoma japonicum/inmunología , Esquistosomiasis Japónica/inmunología , Vacunas Atenuadas , Animales , Antígenos Helmínticos/genética , Antígenos Helmínticos/metabolismo , Rayos gamma , Proteínas del Helminto/genética , Proteínas del Helminto/inmunología , Proteínas del Helminto/metabolismo , Inmunoglobulina G/inmunología , Conejos , Schistosoma japonicum/crecimiento & desarrollo , Schistosoma japonicum/efectos de la radiación , Esquistosomiasis Japónica/parasitología , Esquistosomiasis Japónica/prevención & control , Porcinos , Porcinos Enanos , Vacunas Atenuadas/inmunología , Vacunas de ADN/inmunología
15.
Jpn J Infect Dis ; 62(1): 37-45, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19168957

RESUMEN

To investigate the potential applicability of mucosal vaccines against mucosa-unrelated pathogens, a non-parenteral vaccination approach was taken as a prophylactic strategy against mosquito-borne Japanese encephalitis virus (JEV). Intranasal (i.n.) immunization with a mouse brain-derived formalin-inactivated JE vaccine induced a robust virus-neutralizing antibody in mice, and this induction was augmented by co-administration with cholera toxin (CT) and pertussis toxin, but not with killed Bordetella pertussis. The antibody response induced by the i.n. administration of the JE vaccine with bacterial toxins was comparable in intensity to that induced by a parenteral immunization regime, and the former was considerably more effective in terms of delayed-type hypersensitivity and local antibody response. In addition, the adjuvant effects of bacterial toxins were much more prominent for the mucosal than the parenteral route. Two other non-invasive routes, oral and transcutaneous administration, were examined, but the i.n. route was by far the most effective. Finally, the vaccine efficacy of a chimeric fusion protein between the B subunit of CT and the JEV envelope protein showed some promise for the development of non-invasive JE vaccine. Our results suggest that the mucosal vaccination approach is feasible for a non-mucosal pathogen such as JEV, but that the adjuvant, carrier molecule, and administration route must be optimized for construction of an effective vaccine platform.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/prevención & control , Vacunas contra la Encefalitis Japonesa/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Toxina del Cólera/administración & dosificación , Toxina del Cólera/inmunología , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/virología , Ensayo de Inmunoadsorción Enzimática , Inmunización , Inmunización Secundaria , Vacunas contra la Encefalitis Japonesa/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Mucosa Nasal/inmunología , Pruebas de Neutralización , Toxina del Pertussis/administración & dosificación , Toxina del Pertussis/inmunología , Proteínas Recombinantes/inmunología , Vacunación
16.
J Vet Med Sci ; 71(7): 995-1000, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19652493

RESUMEN

Plants are attractive vaccine production and oral delivery systems. Cereals are excellent candidate for edible vaccines, which can express and store high levels of proteins for extended periods of time without degradation. In this study, we produced a 14-kDa protective surface antigen of Ascaris suum L3 larvae and its fusion chimera with a mucosal carrier molecule cholera toxin B subunit (CTB) in rice (Oryza sativa L.) under the control of the endosperm-specific glutelin-B promoter. We found that the recombinant protein expression levels reached 1.5 microg per seed, a comparably high amount as compared to previously reported transgenic rice expression experiments. Potentials of transgenic rice plants as a source of oral vaccines against swine roundworm are discussed.


Asunto(s)
Ascaris suum/metabolismo , Toxina del Cólera/metabolismo , Oryza/genética , Semillas/metabolismo , Animales , Ingeniería Genética , Oryza/metabolismo , Plantas Modificadas Genéticamente , Proteínas Recombinantes
17.
Infect Genet Evol ; 75: 103964, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31302241

RESUMEN

The receptor for advanced glycation end products (RAGE) recognizes Ca++-binding proteins, such as members of the S100 protein family released by dead or devitalized tissues, and plays an important role in inflammatory responses. We recently identified the Ca++-binding protein, venestatin, secreted from the rodent parasitic nematode, Strongyloides venezuelensis. We herein characterized recombinant venestatin, which is abundantly produced by the silkworm-baculovirus expression system (silkworm-BES), particularly in its interaction with RAGE. Venestatin from silkworm-BES possessed a binding capacity with Ca++ ions and vaccine immunogenicity against S. venezuelensis larvae in mice, which is similar to venestatin produced by the E. coli expression system (EES). Venestatin from silkworm-BES had a higher affinity for human recombinant RAGE than that from EES, and their affinities were Ca++-dependent. RAGE in the mouse lung co-immunoprecipitated with venestatin from silkworm-BES administered intranasally, indicating that it bound endogenous mouse RAGE. The present results suggest that venestatin from silkworm-BES affects RAGE-mediated pathological processes.


Asunto(s)
Proteínas del Helminto/metabolismo , Strongyloides/genética , Animales , Baculoviridae , Bombyx , Calcio/metabolismo , Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Proteínas del Helminto/genética , Humanos , Larva/metabolismo , Unión Proteica
18.
Exp Anim ; 57(1): 1-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18256513

RESUMEN

The mucosal immunization method is a needle-free alternative way of vaccination. This study evaluated the efficacy of mucosal immunization for rabies. Mice were intranasally administered five times with inactivated and concentrated rabies virus antigen (CRV) supplemented with or without cholera toxin (CT). The anti-rabies virus antibody titer of mice intranasally immunized with CRV plus CT (CRV/CT) was comparable to that of mice intraperitoneally immunized twice with the same amount of CRV. Virus neutralizing (VNA) titers of mice immunized intranasally with CRV/CT were slightly lower than those of intraperitoneally immunized mice. Both anti-rabies virus ELISA antibody and VNA titers of mice immunized with CRV without CT were significantly lower than those of mice immunized with CRV/CT. In mice intranasally immunized with CRV/CT, and intraperitoneally immunized mice, high levels of IgG(2a) antibody were detected, suggesting the activation of Th1-driven cellular immunity by the two ways of immunization. All immunized mice were challenged intracerebrally with a lethal dose of virulent rabies virus CVS strain. The survival rates of mice immunized with CRV/CT and CRV without CT were 67% and 17%, respectively, while the rate of intraperitoneally immunized mice was 100%. Antigen-specific whole IgG and IgG(2a), and VNA titers of survived mice were significantly higher than those of dead mice at the challenge day. These data suggest the possibility of intranasal immunization with inactivated antigen as a rabies vaccination strategy and the importance of a mucosal adjuvant such as CT.


Asunto(s)
Inmunización/métodos , Ratones , Vacunas Antirrábicas/administración & dosificación , Rabia/prevención & control , Enfermedades de los Roedores/prevención & control , Administración Intranasal , Animales , Anticuerpos Antivirales/sangre , Toxina del Cólera/administración & dosificación , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunoglobulina G/sangre , Inyecciones Intraperitoneales , Ratones Endogámicos BALB C , Mucosa Nasal , Vacunas de Productos Inactivados/administración & dosificación
19.
Vaccine ; 34(10): 1268-74, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26828455

RESUMEN

The cholera toxin B subunit (CTB) is secreted in its pentameric form from Escherichia coli if its leader peptide is replaced with one of E. coli origin. However, the secretion of the pentamer is generally severely impaired when the molecule is mutated or fused to a foreign peptide. Therefore, we attempted to regenerate pentameric CTB from the inclusion bodies (IBs) of E. coli. Stepwise dialysis of the IBs solubilized in guanidine hydrochloride predominantly generated soluble high-molecular-mass (HMM) aggregates and only a small fraction of pentamer. Three methods to reassemble homogeneous pentameric molecules were evaluated: (i) using a pentameric coiled-coil fusion partner, expecting it to function as an assembly core; (ii) optimizing the protein concentration during refolding; and (iii) eliminating contaminants before refolding. Coiled-coil fusion had some effect, but substantial amounts of HMM aggregates were still generated. Varying the protein concentration from 0.05 mg/mL to 5mg/mL had almost no effect. In contrast, eliminating the contaminants before refolding had a robust effect, and only the pentamer was regenerated, with no detectable HMM aggregates. Surprisingly, the protein concentration at refolding was up to 5mg/mL when the contaminants were removed, with no adverse effects on refolding. The regenerated pentamer was indistinguishable in its biochemical and immunological characteristics from CTB secreted from E. coli or choleragenoid from Vibrio cholerae. This study provides a simple but very efficient strategy for pentamerizing CTB with a highly homogeneous molecular conformation, with which it may be feasible to engineer CTB derivatives and CTB fusion antigens.


Asunto(s)
Toxina del Cólera/química , Cuerpos de Inclusión/química , Pliegue de Proteína , Animales , Cromatografía en Gel , Escherichia coli/metabolismo , Femenino , Ratones Endogámicos BALB C , Señales de Clasificación de Proteína , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/química
20.
Vaccine ; 34(27): 3184-3190, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27105561

RESUMEN

Egg-drop syndrome (EDS) virus is an avian adenovirus that causes a sudden drop in egg production and in the quality of the eggs when it infects chickens, leading to substantial economic losses in the poultry industry. Inactivated EDS vaccines produced in embryonated duck eggs or cell culture systems are available for the prophylaxis of EDS. However, recombinant subunit vaccines that are efficacious and inexpensive are a desirable alternative. In this study, we engineered chimeric fusion proteins in which the trimeric fiber knob domain lacking the triple ß-spiral motif in the fiber shaft region was genetically fused to trimeric coiled coils, such as those of the engineered form of the GCN4 leucine zipper peptide or chicken cartilage matrix protein (CMP). The fusion proteins were expressed predominantly as soluble trimeric proteins in Escherichia coli at levels of 15-80mg/L of bacterial culture. The single immunization of chickens with the purified fusion proteins, at a dose equivalent to 10µg of the knob moiety, elicited serum antibodies with high hemagglutination inhibition (HI) activities, similar to those induced by an inactivated EDS vaccine. A dose-response analysis indicated that a single immunization with as little as 1µg of the knob moiety of the CMP-knob fusion protein was as effective as the inactivated vaccine in inducing antibodies with HI activity. The immunization of laying hens had no apparent adverse effects on egg production and effectively prevented clinical symptoms of EDS when the chickens were challenged with pathogenic EDS virus. This study demonstrates that the knob domain lacking the shaft sequence but fused to a trimeric coiled coil is a promising candidate subunit vaccine for the prophylaxis of EDS in chickens.


Asunto(s)
Infecciones por Adenoviridae/veterinaria , Proteínas de la Cápside/inmunología , Pollos/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunas Virales/inmunología , Infecciones por Adenoviridae/prevención & control , Animales , Anticuerpos Antivirales/sangre , Aviadenovirus , Huevos , Femenino , Pruebas de Inhibición de Hemaglutinación , Enfermedades de las Aves de Corral/virología , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/inmunología , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda