RESUMEN
Aggregation of the tau protein is thought to be responsible for the neurodegeneration and subsequent functional impairments in diseases that are collectively named tauopathies. Alzheimer's disease is the most common tauopathy, but the group consists of over 20 different diseases, many of which have tau pathology as their primary feature. The development of tau therapies has mainly focused on preventing the formation of and/or clearing these aggregates. Of these, immunotherapies that aim to either elicit endogenous tau antibodies or deliver exogenous ones are the most common approach in clinical trials. While their mechanism of action can involve several pathways, both extra- and intracellular, pharmaceutical companies have primarily focused on antibody-mediated clearance of extracellular tau. As we have pointed out over the years, this is rather surprising because it is well known that most of pathological tau protein is found intracellularly. It has been repeatedly shown by several groups over the past decades that antibodies can enter neurons and that their cellular uptake can be enhanced by various means, particularly by altering their charge. Here, we will briefly describe the potential extra- and intracellular mechanisms involved in antibody-mediated clearance of tau pathology, discuss these in the context of recent failures of some of the tau antibody trials, and finally provide a brief overview of how the intracellular efficacy of tau antibodies can potentially be further improved by certain modifications that aim to enhance tau clearance via specific intracellular degradation pathways.
Asunto(s)
Enfermedad de Alzheimer , Inmunoterapia , Tauopatías , Proteínas tau , Enfermedad de Alzheimer/metabolismo , Anticuerpos/uso terapéutico , Humanos , Inmunoterapia/métodos , Neuronas/metabolismo , Tauopatías/tratamiento farmacológico , Tauopatías/metabolismo , Tauopatías/patología , Proteínas tau/inmunología , Proteínas tau/metabolismoRESUMEN
To evaluate the efficacy of oral immunization with active kisspeptin DNA vaccine on the expression of hormone receptor mRNA. For this study, ten 56-day-old Hu breed ram lambs were randomly assigned to the treatment and control groups (n = 5). Treatment Experimental group received C500/pKS-asd and the control group received C500/pVAX-asd (aspartate-ß semialdehyde dehydrogenase orally on days 0, 28, and 56, and blood samples were taken at each immunization interval (14-day) and tissues samples were collected at the end of the experimental period (day 98). The collected samples were stored in the refrigerator at -20 °C and liquid nitrogen, respectively, for laboratory examination. Total RNA was extracted from samples using TRIzol reagent and quantitative real-time polymerase chain reaction (QPCR) was used to quantify the levels of KISS1, G protein-coupled receptor-54 (Kiss1r), and gonadotrophin-releasing hormone (GnRH) mRNA in the hypothalamus. Levels of luteinizing hormone receptor (LHR) and luteinizing hormone beta (LHß) mRNA, and follicle-stimulating hormone receptor (FSHR) and follicle-stimulating hormone beta (FSHß) mRNA in the testes and pituitary were analyzed, respectively. Further, gonadotropin-releasing hormone receptor (GnRHR) mRNA expression level in the pituitary was measured. Moreover, the Kiss1r concentration level in the blood was measured using an indirect ELISA. The concentration of Kiss1r in the blood was lower in the treatment group than in the control group (p < 0.05). The levels of testicular FSHR and LHR mRNA were significantly lower in the treatment group (p < 0.05) when compared to the control group. Furthermore, the treatment group's levels of hypothalamic KISS1, Kiss1r, and GnRH mRNA were significantly lower (p < 0.05) than the controls. LH, FSH, and GnRHR mRNA expression in the pituitary were also significantly lower in the treatment group (p < 0.01 and p < 0.05, respectively). These findings imply that oral immunization with active kisspeptin DNA vaccine suppresses hormone receptor mRNA expression in the ram lambs.
Asunto(s)
Kisspeptinas , Vacunas de ADN , Ovinos/genética , Animales , Masculino , Kisspeptinas/genética , Receptores de Kisspeptina-1 , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Hormona Liberadora de Gonadotropina/genética , Oveja Doméstica/genética , Receptores Acoplados a Proteínas G/genética , Receptores de HL/genética , Inmunización/veterinaria , ARN Mensajero/genética , ARN Mensajero/metabolismo , Hormona Folículo Estimulante/genéticaRESUMEN
Coxsackievirus A5 (CV-A5) has recently emerged as a main hand, foot, and mouth disease (HFMD) pathogen. Following a large-scale vaccination campaign against enterovirus 71 (EV-71) in China, the number of HFMD-associated cases with EV-71 was reduced, especially severe and fatal cases. However, the total number of HFMD cases remains high, as HFMD is also caused by other enterovirus serotypes. A multivalent HFMD vaccine containing 4 or 6 antigens of enterovirus serotypes is urgently needed. A formaldehyde-inactivated CV-A5 vaccine derived from Vero cells was used to inoculate newborn Kunming mice on days 3 and 10. The mice were challenged on day 14 with a mouse-adapted CV-A5 strain at a dose that was lethal for 14-day-old suckling mice. Within 14 days postchallenge, groups of mice immunized with three formulations, empty particles (EPs), full particles (FPs), and a mixture of the EP and FP vaccine candidates, all survived, while 100% of the mock-immunized mice died. Neutralizing antibodies (NtAbs) were detected in the sera of immunized mice, and the NtAb levels were correlated with the survival rate of the challenged mice. The virus loads in organs were reduced, and pathological changes and viral protein expression were weak or not observed in the immunized mice compared with those in alum-inoculated control mice. Another interesting finding was the identification of CV-A5 dense particles (DPs), facilitating morphogenesis study. These results demonstrated that the Vero cell-adapted CV-A5 strain is a promising vaccine candidate and could be used as a multivalent HFMD vaccine component in the future.IMPORTANCE The vaccine candidate strain CV-A5 was produced with a high infectivity titer and a high viral particle yield. Three particle forms, empty particles (EPs), full particles (FPs), and dense particles (DPs), were obtained and characterized after purification. The immunogenicities of EP, FP, and the EP and FP mixture were evaluated in mice. Mouse-adapted CV-A5 was generated as a challenge strain to infect 14-day-old mice. An active immunization challenge mouse model was established to evaluate the efficacy of the inactivated vaccine candidate. This animal model mimics vaccination, similar to immune responses of the vaccinated. The animal model also tests protective efficacy in response to the vaccine against the disease. This work is important for the preparation of multivalent vaccines against HFMD caused by different emerging strains.
Asunto(s)
Enterovirus Humano A/inmunología , Enfermedad de Boca, Mano y Pie/prevención & control , Vacunación/métodos , Vacunas Virales/administración & dosificación , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Chlorocebus aethiops , Modelos Animales de Enfermedad , Enfermedad de Boca, Mano y Pie/virología , Ratones , Serogrupo , Vacunas Combinadas/administración & dosificación , Vacunas Combinadas/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Células Vero , Carga Viral , Vacunas Virales/inmunología , Virión/inmunologíaRESUMEN
Incidence of cancer in the epididymis is very rare. It is proposed that proteins specific to this organ may contribute to this unique property. We previously demonstrated that siRNA-mediated knockdown of SPAG11A mRNA resulted in increased proliferation of epididymal epithelial cells, whereas overexpression of this gene caused reduced proliferation in immortalized cell lines. In this study, we evaluated the oncogenesis-related anatomical and transcriptome changes in the epididymis of SPAG11A-immunized rats challenged with a low dose of diethyl nitrosamine (DEN). DEN treatment or SPAG11A immunization alone did not cause any histopathological changes in the epididymis. Interestingly, indications of oncogenesis were observed in SPAG11A-immunized + DEN-treated rats. Using high throughput sequencing, we observed that 3549 transcripts that were differentially expressed in the caput epididymis of DEN only-treated rats displayed similar differential expression in the caput epididymis of SPAG11A-immunized rats, indicating that the microenvironment that contributes to oncogenesis sets in when SPAG11A protein is ablated. Differential expression of genes that are involved in 10 major cancer related pathways was also analyzed. Majority of the genes related to these pathways that were differentially expressed in the caput epididymis of DEN only-treated rats also showed similar pattern in the caput epididymis of SPAG11A-immunized rats. For the first time, results of our study demonstrate that ablation of SPAG11A by active immunization renders the epididymis susceptible to oncogenesis and that this protein may be one of the factors that contributes to the rarity of epididymal cancer.
Asunto(s)
Epidídimo , beta-Defensinas , Animales , Carcinogénesis/metabolismo , Epidídimo/metabolismo , Masculino , Proteínas/metabolismo , Ratas , Espermatozoides/metabolismo , Microambiente Tumoral , Vacunación , beta-Defensinas/genéticaRESUMEN
AIM: Unexplained infertility is a major burden for couples who want to have children. Lymphocyte immunotherapy (LIT) could be a therapeutic help for these couples. Although LIT has been carried out for decades, the data on the success of therapy are still controversial and there is hardly information on possible adverse drug reactions. METHODS: In this study, we used a questionnaire to determine the frequency of local and systemic adverse drug reactions in our patients who were treated with LIT between 2017 and 2020 (n = 302). In addition, we asked about pregnancies and/or live births after LIT in a 2-year follow-up (n = 140). RESULTS: Most of the patients reported the occurrence of mild local adverse drug reactions in a period of less than 4 weeks: Over 75% reported moderate erythema, itching or swelling, over 10% erythema, itching or swelling as more pronounced adverse drug reaction. Blistering was specified in 10% of the cases. Serious adverse drug reactions or adverse events were not described. In the follow-up, 69% of our patients stated a pregnancy after LIT, and 50% a life birth. CONCLUSIONS: Overall, LIT represents a well-tolerated therapy for couples with unexplained infertility, however, more evidence is needed on the benefits.
Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Infertilidad , Niño , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/terapia , Femenino , Humanos , Inmunoterapia/efectos adversos , Infertilidad/terapia , Nacimiento Vivo , Linfocitos , Embarazo , Índice de Embarazo , Prurito , Estudios RetrospectivosRESUMEN
Transmissible spongiform encephalopathies are incurable neurodegenerative diseases, associated with the conversion of the physiological prion protein to its disease-associated counterpart. Even though immunization against transmissible spongiform encephalopathies has shown great potential, immune tolerance effects impede the use of active immunization protocols for successful prophylaxis. In this study, we evaluate the use of trypanosomes as biological platforms for the presentation of a prion antigenic peptide to the host immune system. Using the engineered trypanosomes in an immunization protocol without the use of adjuvants led to the development of a humoral immune response against the prion protein in wild type mice, without the appearance of adverse reactions. The immune reaction elicited with this protocol displayed in vitro therapeutic potential and was further evaluated in a bioassay where immunized mice were partially protected in a representative murine model of prion diseases. Further studies are underway to better characterize the immune reaction and optimize the immunization protocol.
Asunto(s)
Enfermedades por Prión , Priones , Trypanosoma , Animales , Inmunización , Ratones , Enfermedades por Prión/prevención & control , Proteínas Priónicas , Priones/genética , VacunaciónRESUMEN
BACKGROUND: Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis is a recently discovered autoimmune syndrome associated with psychosis, dyskinesia, and seizures. However, the underlying mechanisms of this disease remain unclear, in part because of a lack of suitable animal models. METHODS: This study describes a novel female C57BL/6 mouse model of anti-NMDAR encephalitis that was induced by active immunization against NMDARs using an amino terminal domain (ATD) peptide from the GluN1 subunit (GluN1356-385). RESULTS: Twelve weeks after immunization, the immunized mice showed significant memory loss. Furthermore, antibodies from the cerebrospinal fluid of immunized mice decreased the surface NMDAR cluster density in hippocampal neurons which was similar to the effect induced by the anti-NMDAR encephalitis patients' antibodies. Immunization also impaired long-term potentiation at Schaffer collateral-CA1 synapses and reduced NMDAR-induced calcium influx. CONCLUSION: We established a novel anti-NMDAR encephalitis model using active immunization with peptide GluN1356-385 targeting the ATD of GluN1. This novel model may allow further research into the pathogenesis of anti-NMDAR encephalitis and aid in the development of new therapies for this disease.
Asunto(s)
Encefalitis Antirreceptor N-Metil-D-Aspartato/inducido químicamente , Proteínas del Tejido Nervioso/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Receptores de N-Metil-D-Aspartato/administración & dosificación , Vacunación/efectos adversos , Secuencia de Aminoácidos , Animales , Encefalitis Antirreceptor N-Metil-D-Aspartato/genética , Encefalitis Antirreceptor N-Metil-D-Aspartato/inmunología , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Células Cultivadas , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Ratas , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/inmunología , Vacunación/métodosRESUMEN
Unintentional overdose deaths related to opioids and psychostimulants have increased in prevalence due to the adulteration of these drugs with fentanyl. Synergistic effects between illicit compounds and fentanyl cause aggravated respiratory depression, leading to inadvertent fatalities. Traditional small-molecule therapies implemented in the expanding opioid epidemic present numerous problems since they interact with the same opioid receptors in the brain as the abused drugs. In this study, we report an optimized dual hapten for use as an immunopharmacotherapeutic tool in order to develop antibodies capable of binding to fentanyl-contaminated heroin in the periphery, thus impeding the drugs' psychoactive effects on the central nervous system. This vaccine produced antibodies with nanomolar affinities and effectively blocked opioid analgesic effects elicited by adulterated heroin. These findings provide further insight into the development of chemically contiguous haptens for broad-spectrum immunopharmacotherapies against opioid use disorders.
Asunto(s)
Sobredosis de Droga/prevención & control , Fentanilo/inmunología , Haptenos/inmunología , Heroína/efectos adversos , Heroína/química , Vacunas/inmunología , Animales , Contaminación de Medicamentos , Sobredosis de Droga/mortalidad , Fentanilo/efectos adversos , Fentanilo/química , Humanos , Ratones , Trastornos Relacionados con OpioidesRESUMEN
Drug addiction is a major worldwide medical and social problem.Cocaine,nicotine,methamphetamine,heroin and other psychoactive substances,with small molecular weight,can easily cross the blood-brain barrier and eventually lead to addiction and other serious neuropsychological damage.There is no effective cure for addiction currently.The drug-antibody complex formed on the basis of active or passive immunotherapy could not cross the blood-brain barrier,which reduces the concentration of the free active drug and prevents its distribution in the brain,thereby weakening the drug addiction-related reward effects.It provides a promising way for the treatment of drug addiction.This article reviews the progress of immunotherapy against psychoactive substances such as cocaine,nicotine,methamphetamine and heroin in the past 50 years from the aspects of active immunity,passive immunity,drug metabolism-related enzymes,adjuvants and so on.The goal is to provide some ideas for the development of agents for the treatment of psychoactive substance addiction.
Asunto(s)
Cocaína , Metanfetamina , Trastornos Relacionados con Sustancias , Humanos , Inmunoterapia , Nicotina , Trastornos Relacionados con Sustancias/terapiaRESUMEN
Multiple system atrophy (MSA) is a rare and fatal neurodegenerative disease with limited symptomatic treatment options. Aggregation of α-synuclein in oligodendrocytes is believed to be a central mechanism of the neurodegenerative process. PD01A and PD03A are 2 novel therapeutic vaccine candidates containing short peptides as antigenic moieties that are designed to induce a sustained antibody response, specifically targeting pathogenic assemblies of α-synuclein. The objectives of the current study were to evaluate primarily the safety and tolerability of PD01A and PD03A in patients with early MSA. Thirty patients (11 women) were randomized to receive 5 subcutaneous injections of either PD01A (n = 12), PD03A (n = 12), or placebo (n = 6) in this patient- and examiner-blinded, placebo-controlled, 52-week phase 1 clinical trial (ClinicalTrial.gov identifier: NCT02270489). Immunogenicity and clinical scores were assessed as secondary objectives. Twenty-nine patients reported a total of 595 treatment-emergent adverse events (mild or moderate, n = 555; severe, n = 40). Treatment-related adverse events included 190 injection-site reactions typically observed in vaccination trials with similar per-subject incidence in the treatment groups over time. Sustained IgG titers were observed in the PD01A-treated group, and 89% of treated patients developed a PD01-specific antibody response after receiving all injections. Induced antibodies displayed clear reactivity to the α-synuclein target epitope. Titers and antibody responder rate (58%) were lower in the PD03A-treated group. In conclusion, both PD01A and PD03A were safe and well tolerated. PD01A triggered a rapid and long-lasting antibody response that specifically targeted the α-synuclein epitope. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Asunto(s)
Atrofia de Múltiples Sistemas , Enfermedad de Parkinson , Femenino , Humanos , Masculino , Atrofia de Múltiples Sistemas/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , Péptidos , Vacunación , alfa-SinucleínaRESUMEN
FGF-2 accumulates in many tumor tissues and is closely related to the development of tumor angiogenesis and the immunosuppressive microenvironment. This study aimed to investigate whether active immunization against FGF-2 could modify antitumor immunity and enhance the efficacy of an HPV16 E7-specific therapeutic vaccine. Combined immunization targeting both FGF-2 and E7 significantly suppressed tumor growth, which was accompanied by significantly increased levels of IFN-γ-expressing splenocytes and effector CD8 T cells and decreased levels of immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells(MDSCs) in both the spleen and tumor; in addition, the levels of FGF-2 and neovascularization in tumors were decreased in the mice receiving the combined immunization, and tumor cell apoptosis was promoted. The combination of an HPV16 E7-specific vaccine and active immunization against FGF-2 significantly enhances antitumor immune responses in mice with TC-1 tumors, indicating a promising strategy for tumor immunotherapy.
Asunto(s)
Vacunas contra el Cáncer/farmacología , Factor 2 de Crecimiento de Fibroblastos/inmunología , Neovascularización Patológica/inmunología , Proteínas E7 de Papillomavirus/inmunología , Vacunas contra Papillomavirus/farmacología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Inmunoterapia , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Neovascularización Patológica/virología , Proteínas E7 de Papillomavirus/antagonistas & inhibidores , Proteínas E7 de Papillomavirus/genética , Vacunas contra Papillomavirus/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , VacunaciónRESUMEN
Active and passive immunization have been used to treat human disease for hundreds of years and improvements in technology and knowledge is only increasing the number of therapeutic applications. The current and future use of immunization to treat neurodegenerative diseases are briefly described herein to serve as an introduction to this special issue.
Asunto(s)
Inmunización Pasiva/métodos , Enfermedades Neurodegenerativas/terapia , Vacunación/métodos , HumanosRESUMEN
The types of treatments and interventions being developed for chronic neurodegenerative disorders have expanded considerably in recent years. In addition to the variety of targets being pursued, strategies have moved from symptom management to more directed disease-modifying approaches. Among them are antibody-based therapies, which are not only being evaluated for a range of tauopathies and synucleinopathies, but are also emerging as a potential application for monogenic disorders of the central nervous system (CNS), including Huntington's disease (HD). Despite the excitement around the early trial data of anti-sense oligonucleotides (ASO) treatment for such disorders, antibody therapies may hold the key to tackling another aspect of the disease that could be critical to its pathogenesis. While gene-based methodologies are designed to lower, predominantly within cellular elements, mutant huntingtin protein (mHtt) - the genetic product of HD - the pathological protein is abundant in free forms and in several compartments including the cerebrospinal fluid, the plasma and the extracellular matrix. With accumulating evidence for the spreading and seeding capacities of mHtt, it may indeed be essential to target the protein both intracellularly and extracellularly. Therefore, free forms of mHtt not only represents an ideal target for antibodies, but one that needs to be addressed if meaningful and maximal clinical benefits are to be expected. This review explores the potential use of antibody-based therapies to treat HD, including the rationale for this approach as well as the pre-clinical data supporting it. The potential challenges that will need to be considered if such route is to be pursued clinically are also discussed.
Asunto(s)
Anticuerpos/uso terapéutico , Enfermedad de Huntington/terapia , Inmunización/métodos , Inmunización/tendencias , Animales , Humanos , Proteína Huntingtina/antagonistas & inhibidoresRESUMEN
BACKGROUND: Active immunotherapy targeting amyloid-ß (Aß) is a promising treatment for Alzheimer's disease (AD). Numerous preclinical studies and clinical trials demonstrated that a safe and effective AD vaccine should induce high titers of anti-Aß antibodies while avoiding the activation of T cells specific to Aß. RESULTS: An untagged Aß1-6 chimeric protein vaccine against AD based on norovirus (NoV) P particle was expressed in Escherichia coli and obtained by sequential chromatography. Analysis of protein characteristics showed that the untagged Aß1-6 chimeric protein expressed in soluble form exhibited the highest particle homogeneity, with highest purity and minimal host cell protein (HCP) and residual DNA content. Importantly, the untagged Aß1-6 chimeric soluble protein could induce the strongest Aß-specific humoral immune responses without activation of harmful Aß-specific T cells in mice. CONCLUSIONS: The untagged Aß1-6 chimeric protein vaccine is safe and highly immunogenic. Further research will determine the efficacy in cognitive improvement and disease progression delay.
Asunto(s)
Vacunas contra el Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Anticuerpos/inmunología , Vectores Genéticos , Norovirus , Proteínas Recombinantes de Fusión , Enfermedad de Alzheimer/terapia , Vacunas contra el Alzheimer/administración & dosificación , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/aislamiento & purificación , Animales , Especificidad de Anticuerpos/inmunología , Modelos Animales de Enfermedad , Inmunización , Ratones , Norovirus/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismoRESUMEN
The objective of this study was to investigate effects of active immunization against Akirin2 on muscle fiber-type composition in pigs. Here we showed that the titer of Akirin2 antibody in pigs immunized with porcine Akirin2 (pAkirin2) was significantly increased. Active immunization against pAkirin2 decreased succinic dehydrogenase and malate dehydrogenase activities and increased lactate dehydrogenase activity in the longissimus dorsi muscle of pigs. Active immunization against pAkirin2 significantly decreased MyHC I and MyHC IIa mRNA expressions and MyHC I protein expression and increased mRNA expressions of MyHC IIb as well as protein expressions of MyHC IIb and fast-MyHC. mRNA expressions of nuclear factors of activated T cells c1 (NFATc1), transcriptional coactivator PPARγ coactivator-1α, myocyte enhancer factor 2C, and modulatory calcineurin interacting protein 1 exon 4 isform were also notably decreased by active immunization against pAkirin2. Together, our data imply that active immunization against pAkirin2 may result in a slow to fast fiber-type shift in pigs, and which may be mediated by suppression of the calcineurin/NFATc1 signaling pathway.
Asunto(s)
Regulación de la Expresión Génica , Fibras Musculares Esqueléticas/metabolismo , Proteínas Represoras/metabolismo , Porcinos/fisiología , Vacunación/veterinaria , Animales , Calcineurina/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Malato Deshidrogenasa/metabolismo , Masculino , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Lenta/metabolismo , Factores de Transcripción NFATC/metabolismo , ARN Mensajero/metabolismo , Distribución Aleatoria , Proteínas Represoras/genética , Succinato Deshidrogenasa/metabolismoRESUMEN
Setting up an animal model by using active immunization methods is a common means of studying immune-related diseases or producing antibodies with high titer and high activities. However, the security during the process of pathogen emulsification remains unclear. In a physical examination, we unexpectedly noticed high levels of angiotensin II type 1 receptor autoantibody (AT1-AA) specific to the immunizing antigen in the sera of some researchers who had participated in setting up active immunization animal models, and we were puzzled about the cause of AT1-AA production. In this study, we intended to investigate whether the emulsified antigen was the source of infection in these researchers, and if so, how to prevent it from occurring. AT1-AA was detected by advanced ELISA method. The participants presented higher levels of AT1-AA compared with non-participants of the same laboratory. This phenomenon remained that some factors during the process of rat model establishment may contribute to AT1-AA production. Animal and glove penetration studies indicated the emulsified antigen infection was attributed to neither aerosol or fur touch nor penetrating through gloves. However, AT1-AA level was largely decreased in the participants after they used an automatic emulsification device. Because of the strong permeability of the adjuvant, we speculated that emulsified antigen might get access to the unprotected skin of the participants accidentally during the immunization process. These results demonstrated that accidental contacts of emulsified antigens may infect researchers during the process of traditional hand-push emulsification, resulting in high specific autoantibody levels, which can be prevented by using appropriate tools.
Asunto(s)
Emulsiones/química , Inmunización/métodos , Administración Cutánea , Adulto , Animales , Antígenos/inmunología , Autoanticuerpos/sangre , Femenino , Humanos , Masculino , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/inmunología , Factores de Riesgo , Adulto JovenRESUMEN
Vaccination therapies constitute potential treatment options in neurodegenerative disorders such as Alzheimer disease or Parkinson disease. While a lot of research has been performed on vaccination against extracellular amyloid ß, the focus recently shifted toward vaccination against the intracellular proteins tau and α-synuclein, with promising results in terms of protein accumulation reduction. In this review, we briefly summarize lessons to be learned from clinical vaccination trials in Alzheimer disease that target amyloid ß. We then focus on tau and α-synuclein. For both proteins, we provide important data on protein immunogenicity, and put them into context with data available from both animals and human vaccination trials targeted at tau and α-synuclein. Together, we give a comprehensive overview about current clinical data, and discuss associated problems.
Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Enfermedad de Parkinson/inmunología , Tauopatías/inmunología , Vacunación , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Enfermedad de Parkinson/prevención & control , Tauopatías/metabolismo , Vacunación/métodos , alfa-Sinucleína/metabolismoRESUMEN
Strategies aimed at reducing cerebral accumulation of the amyloid-ß (Aß) peptides have therapeutic potential in Alzheimer's disease (AD). Aß immunization has proven to be effective at promoting Aß clearance in animal models but adverse effects have hampered its clinical evaluation. The first anti-Aß immunization clinical trial, which assessed a full-length Aß1-42 vaccine, increased the risk of encephalitis most likely because of autoimmune pro-inflammatory T helper 1 (Th1) response against all forms of Aß. Immunization against less abundant but potentially more pathologically relevant Aß products, such as N-terminally-truncated pyroglutamate-3 Aß (AßpE3), could provide efficacy and improve tolerability in Aß immunotherapy. Here, we describe a selective vaccine against AßpE3, which uses the diphtheria toxin mutant CRM197 as carrier protein for epitope presentation. CRM197 is currently used in licensed vaccines and has demonstrated excellent immunogenicity and safety in humans. In mice, our AßpE3:CRM197 vaccine triggered the production of specific anti-AßpE3 antibodies that did not cross-react with Aß1-42, non-cyclized AßE3, or N-terminally-truncated pyroglutamate-11 Aß (AßpE11). AßpE3:CRM197 antiserum strongly labeled AßpE3 in insoluble protein extracts and decorated cortical amyloid plaques in human AD brains. Anti-AßpE3 antibodies were almost exclusively of the IgG1 isotype, suggesting an anti-inflammatory Th2 response bias to the AßpE3:CRM197 vaccine. To the best of our knowledge, this study shows for the first time that CRM197 has potential as a safe and suitable vaccine carrier for active and selective immunization against specific protein sequence modifications or conformations, such as AßpE3.
RESUMEN
OBJECTIVE: The efficiency and safety of vaccine are the most important properties, however, as any medication, it can induce side effects. This prophylactic therapy could be used to prevent the lethal and pathophysiological effects induced after scorpion envenomation. METHODS: In this study, detoxified venom associated to alum adjuvant (V*alum) is used as a vaccine against scorpion venom for immunization of mice. We evaluate the safety and the inflammatory response of this vaccine. We also investigated the protective effect of this formulation against the toxicity of native Androctonus australis hector venom. RESULTS: Results showed no adverse events occurred after immunization of animals. This active immunization of animals did not cause change in vascular permeability, no edema formation in the studied organs. Furthermore, there are no IgE production in sera, nor change in the morphology of the mast cells in skin tissues. However, low inflammatory response triggered by activating the recruitment of eosinophils associated to IL-4 and IL-5 release was observed. All immunized animals are protected from the toxic effects of native venom until 6 LD50 and to 7 LD50 after the second challenge. CONCLUSION: This safe vaccine preparation seems to induce a long-term protection without any risk of deleterious inflammatory response.
Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre/administración & dosificación , Venenos de Escorpión/administración & dosificación , Mordeduras de Serpientes , Animales , Peroxidasa del Eosinófilo/inmunología , Eosinófilos/inmunología , Femenino , Inmunoglobulina E/sangre , Interleucina-4/sangre , Interleucina-5/sangre , Ratones , Venenos de Escorpión/inmunología , VacunaciónRESUMEN
AIM: In vitro Influenzinum induce a cellular change. We present the results of the first study examining the effectiveness of Influenzinum against influenza-like illnesses. METHOD: Retrospective cohort study during winter 2014-2015. After influenza epidemic, a self-assessment questionnaire was offered to patients presenting for a consultation. The primary endpoint was the declaration of an influenza-like illness. The exposed patients (treated by Influenzinum) were matched to two non-exposed patients (untreated) with a propensity score. A conditional logistic model expressed influenza-like illness risk reduction provided by the Influenzinum. RESULTS: The cohort included 3514 patients recruited from 46 general practitioners. After matching, the treated group (n=2041) and the untreated group (n=482) did not differ on variables collected. Influenzinum preventive therapy does not significantly alter the likelihood of influenza-like illness (adjusted odds ratio=0,91 [0,62 to 1,35], p=0,64). CONCLUSION: Influenzinum preventive therapy did not appear effective in preventing influenza-like illness.