Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 14313, 2022 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-35995959

RESUMO

Novel approaches for malaria prophylaxis remain important. Synthetic DNA-encoded monoclonal antibodies (DMAbs) are a promising approach to generate rapid, direct in vivo host-generated mAbs with potential benefits in production simplicity and distribution coupled with genetic engineering. Here, we explore this approach in a malaria challenge model. We engineered germline-reverted DMAbs based on human mAb clones CIS43, 317, and L9 which target a junctional epitope, major repeat, and minor repeat of the Plasmodium falciparum circumsporozoite protein (CSP) respectively. DMAb variants were encoded into a plasmid vector backbone and their expression and binding profiles were characterized. We demonstrate long-term serological expression of DMAb constructs resulting in in vivo efficacy of CIS43 GL and 317 GL in a rigorous mosquito bite mouse challenge model. Additionally, we engineered an Fc modified variant of CIS43 and L9-based DMAbs to ablate binding to C1q to test the impact of complement-dependent Fc function on challenge outcomes. Complement knockout variant DMAbs demonstrated similar protection to that of WT Fc DMAbs supporting the notion that direct binding to the parasite is sufficient for the protection observed. Further investigation of DMAbs for malaria prophylaxis appears of importance.


Assuntos
Anticorpos Monoclonais , Vacinas Antimaláricas , Malária Falciparum , Animais , Anticorpos Antiprotozoários , DNA , Modelos Animais de Doenças , Humanos , Vacinas Antimaláricas/genética , Malária Falciparum/prevenção & controle , Camundongos , Plasmodium falciparum , Proteínas de Protozoários
2.
Cell Rep Med ; 2(10): 100420, 2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34604818

RESUMO

Coronavirus disease 2019 (COVID-19), caused by the SARS-CoV-2 virus, has had a dramatic global impact on public health and social and economic infrastructures. Here, we assess the immunogenicity and anamnestic protective efficacy in rhesus macaques of an intradermal (i.d.)-delivered SARS-CoV-2 spike DNA vaccine, INO-4800, currently being evaluated in clinical trials. Vaccination with INO-4800 induced T cell responses and induced spike antigen and RBD binding antibodies with ADCP and ADCD activity. Sera from the animals neutralized both the D614 and G614 SARS-CoV-2 pseudotype viruses. Several months after vaccination, animals were challenged with SARS-CoV-2 resulting in rapid recall of anti-SARS-CoV-2 spike protein T cell and neutralizing antibody responses. These responses were associated with lower viral loads in the lung. These studies support the immune impact of INO-4800 for inducing both humoral and cellular arms of the adaptive immune system, which are likely important for providing durable protection against COVID-19 disease.


Assuntos
Anticorpos Antivirais/sangue , Vacinas contra COVID-19/administração & dosagem , COVID-19/prevenção & controle , Pulmão/virologia , Linfócitos T/imunologia , Animais , Anticorpos Neutralizantes/sangue , Vacinas contra COVID-19/uso terapêutico , Feminino , Injeções Intradérmicas , Macaca mulatta , Masculino , SARS-CoV-2/imunologia , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/uso terapêutico , Carga Viral
3.
Infect Immun ; 89(10): e0072820, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34152830

RESUMO

Malaria infects millions of people every year, and despite recent advances in controlling disease spread, such as vaccination, it remains a global health concern. The circumsporozoite protein (CSP) has long been acknowledged as a key target in antimalarial immunity. Leveraging the DNA vaccine platform against this formidable pathogen, the following five synthetic DNA vaccines encoding variations of CSP were designed and studied: 3D7, GPI1, ΔGPI, TM, and DD2. Among the single CSP antigen constructs, a range of immunogenicity was observed with ΔGPI generating the most robust immunity. In an intravenous (i.v.) sporozoite challenge, the best protection among vaccinated mice was achieved by ΔGPI, which performed almost as well as the monoclonal antibody 311 (MAb 311) antibody control. Further analyses revealed that ΔGPI develops high-molecular-weight multimers in addition to monomeric CSP. We then compared the immunity generated by ΔGPI versus synDNA mimics for the antimalaria vaccines RTS,S and R21. The anti-CSP antibody responses induced were similar among these three immunogens. T cell responses demonstrated that ΔGPI induced a more focused anti-CSP response. In an infectious mosquito challenge, all three of these constructs generated inhibition of liver-stage infection as well as immunity from blood-stage parasitemia. This study demonstrates that synDNA mimics of complex malaria immunogens can provide substantial protection as can a novel synDNA vaccine ΔGPI.


Assuntos
Imunogenicidade da Vacina/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária/imunologia , Proteínas de Protozoários/imunologia , Vacinas Sintéticas/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/imunologia , Linhagem Celular , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium berghei/imunologia , Plasmodium falciparum/imunologia , Esporozoítos/imunologia , Vacinação/métodos
4.
iScience ; 24(7): 102699, 2021 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-34124612

RESUMO

More than 100 million people have been infected with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Common laboratory mice are not susceptible to wild-type SARS-CoV-2 infection, challenging the development and testing of effective interventions. Here, we describe the development and testing of a mouse model for SARS-CoV-2 infection based on transduction of the respiratory tract of laboratory mice with an adeno-associated virus vector (AAV6) expressing human ACE-2 (AAV6.2FF-hACE2). We validated this model using a previously described synthetic DNA vaccine plasmid, INO-4800 (pS). Intranasal instillation of AAV6.2FF-hACE2 resulted in robust hACE2 expression in the respiratory tract. pS induced robust cellular and humoral responses. Vaccinated animals were challenged with 105 TCID50 SARS-CoV-2 (hCoV-19/Canada/ON-VIDO-01/2020) and euthanized four days post-challenge to assess viral load. One immunization resulted in 50% protection and two immunizations were completely protective. Overall, the AAV6.2FF-hACE2 mouse transduction model represents an easily accessible, genetically diverse mouse model for wild-type SARS-CoV-2 infection and preclinical evaluation of potential interventions.

5.
Front Cell Infect Microbiol ; 11: 804186, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35111697

RESUMO

Upon transmission to the human host, Plasmodium sporozoites exit the skin, are taken up by the blood stream, and then travel to the liver where they infect and significantly modify a single hepatocyte. Low infection rates within the liver have made proteomic studies of infected hepatocytes challenging, particularly in vivo, and existing studies have been largely unable to consider how protein and phosphoprotein differences are altered at different spatial locations within the heterogeneous liver. Using digital spatial profiling, we characterized changes in host signaling during Plasmodium yoelii infection in vivo without disrupting the liver tissue. Moreover, we measured alterations in protein expression around infected hepatocytes and identified a subset of CD163+ Kupffer cells that migrate towards infected cells during infection. These data offer the first insight into the heterogeneous microenvironment that surrounds the infected hepatocyte and provide insights into how the parasite may alter its milieu to influence its survival and modulate immunity.


Assuntos
Malária , Plasmodium , Animais , Humanos , Fígado/parasitologia , Malária/parasitologia , Proteômica , Esporozoítos
6.
Nat Commun ; 11(1): 2601, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32433465

RESUMO

The coronavirus family member, SARS-CoV-2 has been identified as the causal agent for the pandemic viral pneumonia disease, COVID-19. At this time, no vaccine is available to control further dissemination of the disease. We have previously engineered a synthetic DNA vaccine targeting the MERS coronavirus Spike (S) protein, the major surface antigen of coronaviruses, which is currently in clinical study. Here we build on this prior experience to generate a synthetic DNA-based vaccine candidate targeting SARS-CoV-2 S protein. The engineered construct, INO-4800, results in robust expression of the S protein in vitro. Following immunization of mice and guinea pigs with INO-4800 we measure antigen-specific T cell responses, functional antibodies which neutralize the SARS-CoV-2 infection and block Spike protein binding to the ACE2 receptor, and biodistribution of SARS-CoV-2 targeting antibodies to the lungs. This preliminary dataset identifies INO-4800 as a potential COVID-19 vaccine candidate, supporting further translational study.


Assuntos
Antígenos Virais/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Enzima de Conversão de Angiotensina 2 , Animais , Anticorpos Neutralizantes/imunologia , Antígenos Virais/química , Vacinas contra COVID-19 , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Mapeamento de Epitopos , Cobaias , Imunidade Humoral , Imunoglobulina G/imunologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Coronavírus da Síndrome Respiratória do Oriente Médio , Modelos Animais , Peptidil Dipeptidase A/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Vacinas Virais/química
7.
Parasitol Res ; 119(4): 1201-1208, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32036440

RESUMO

Knowledge of vector species composition and monitoring their change over time is critical to evaluate malaria transmission and assess control interventions. This is especially important in countries such as Botswana, where malaria transmission is subjected to fluctuations due to climate variability. Another important aspect that impacts vector populations is the insecticide resistance. In order to assess species composition and the presence of mutations associated with insecticide resistance, Anopheles specimens from larval samplings and indoor pyrethrum spray sheet collections were analysed. A total of 349 Anopheles were screened by morphology and PCR as belonging to the An. gambiae complex and An. funestus group. Specimens were subsequently analysed for human blood meal and Plasmodium index. Finally, knock-down resistance polymorphisms were assessed. Anopheles arabiensis accounted for the majority of specimens collected through larval (96.7%) and pyrethrum spray sheet collection (87.4%) across all sampling sites, and this species was the only one found positive for human blood and for P. falciparum. Other Anopheles species were collected in small numbers by pyrethrum spray catches, namely An. quadriannulatus, An. longipalpis type C and An. parensis. The authors speculate on changing climate patterns and their possible impact on species composition. The kdr assay revealed that Anopheles mosquitoes were homozygous wild type for both L1014F (kdr-w) and L1014S (kdr-e) mutations. These results highlight the unique vectorial role of An. arabiensis in Botswana and indicated that even with prolonged use of pyrethroids and DDT, the mosquito population has not developed kdr mutations, despite some in vivo evidence of insecticide resistance.


Assuntos
Anopheles/efeitos dos fármacos , Resistência a Inseticidas , Inseticidas/farmacologia , Piretrinas/farmacologia , Animais , Botsuana , Feminino , Humanos , Resistência a Inseticidas/genética , Larva/efeitos dos fármacos , Malária/transmissão , Mosquitos Vetores/efeitos dos fármacos , Mosquitos Vetores/genética , Mutação , Reação em Cadeia da Polimerase
8.
Vaccines (Basel) ; 8(1)2020 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-31936739

RESUMO

The need for a malaria vaccine is indisputable. A single vaccine for Plasmodium pre-erythrocytic stages targeting the major sporozoite antigen circumsporozoite protein (CSP) has had partial success. Additionally, CD8+ T cells targeting liver-stage (LS) antigens induced by live attenuated sporozoite vaccines were associated with protection in human challenge experiments. To further evaluate protection mediated by LS antigens, we focused on exported pre-erythrocytic proteins (exported protein 1 (EXP1), profilin (PFN), exported protein 2 (EXP2), inhibitor of cysteine proteases (ICP), transmembrane protein 21 (TMP21), and upregulated in infective sporozoites-3 (UIS3)) expressed in all Plasmodium species and designed optimized, synthetic DNA (synDNA) immunogens. SynDNA antigen cocktails were tested with and without the molecular adjuvant plasmid IL-33. Immunized animals developed robust T cell responses including induction of antigen-specific liver-localized CD8+ T cells, which were enhanced by the co-delivery of plasmid IL-33. In total, 100% of mice in adjuvanted groups and 71%-88% in non-adjuvanted groups were protected from blood-stage disease following Plasmodium yoelii sporozoite challenge. This study supports the potential of synDNA LS antigens as vaccine components for malaria parasite infection.

9.
Elife ; 72018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30192230

RESUMO

The TP53 tumor-suppressor gene is mutated in >50% of human tumors and Li-Fraumeni patients with germ line inactivation are predisposed to developing cancer. Here, we generated tp53 deleted zebrafish that spontaneously develop malignant peripheral nerve-sheath tumors, angiosarcomas, germ cell tumors, and an aggressive Natural Killer cell-like leukemia for which no animal model has been developed. Because the tp53 deletion was generated in syngeneic zebrafish, engraftment of fluorescent-labeled tumors could be dynamically visualized over time. Importantly, engrafted tumors shared gene expression signatures with predicted cells of origin in human tissue. Finally, we showed that tp53del/del enhanced invasion and metastasis in kRASG12D-induced embryonal rhabdomyosarcoma (ERMS), but did not alter the overall frequency of cancer stem cells, suggesting novel pro-metastatic roles for TP53 loss-of-function in human muscle tumors. In summary, we have developed a Li-Fraumeni zebrafish model that is amenable to large-scale transplantation and direct visualization of tumor growth in live animals.


Assuntos
Rabdomiossarcoma Embrionário/metabolismo , Rabdomiossarcoma Embrionário/patologia , Proteína Supressora de Tumor p53/deficiência , Peixe-Zebra/metabolismo , Animais , Contagem de Células , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Hemangiossarcoma/metabolismo , Hemangiossarcoma/patologia , Homozigoto , Leucemia/metabolismo , Leucemia/patologia , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias Embrionárias de Células Germinativas/metabolismo , Neoplasias Embrionárias de Células Germinativas/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Rabdomiossarcoma Embrionário/genética , Análise de Sobrevida , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra/genética
10.
Virulence ; 8(8): 1695-1707, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-28614673

RESUMO

White nose syndrome (WNS) is caused by the psychrophilic fungus Pseudogymnoascus destructans that can grow in the environment saprotrophically or parasitically by infecting hibernating bats. Infections are pathological in many species of North American bats, disrupting hibernation and causing mortality. To determine what fungal pathways are involved in infection of living tissue, we examined fungal gene expression using RNA-Seq. We compared P. destructans gene expression when grown in culture to that during infection of a North American bat species, Myotis lucifugus, that shows high WNS mortality. Cultured P. destructans was grown at 10 to 14 C and P. destructans growing in vivo was presumably exposed to temperatures ranging from 4 to 8 C during torpor and up to 37 C during periodic arousals. We found that when P. destructans is causing WNS, the most significant differentially expressed genes were involved in heat shock responses, cell wall remodeling, and micronutrient acquisition. These results indicate that this fungal pathogen responds to host-pathogen interactions by regulating gene expression in ways that may contribute to evasion of host responses. Alterations in fungal cell wall structures could allow P. destructans to avoid detection by host pattern recognition receptors and antibody responses. This study has also identified several fungal pathways upregulated during WNS infection that may be candidates for mitigating infection pathology. By identifying host-specific pathogen responses, these observations have important implications for host-pathogen evolutionary relationships in WNS and other fungal diseases.


Assuntos
Ascomicetos/genética , Quirópteros/microbiologia , Proteínas Fúngicas/genética , Micoses/veterinária , Doenças Nasais/veterinária , Nariz/microbiologia , Transcriptoma , Animais , Ascomicetos/classificação , Ascomicetos/isolamento & purificação , Ascomicetos/fisiologia , Quirópteros/fisiologia , Proteínas Fúngicas/metabolismo , Hibernação , Micoses/microbiologia , Doenças Nasais/microbiologia , Filogenia
11.
Cell Rep ; 19(11): 2304-2318, 2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28614716

RESUMO

Tumor-propagating cells (TPCs) share self-renewal properties with normal stem cells and drive continued tumor growth. However, mechanisms regulating TPC self-renewal are largely unknown, especially in embryonal rhabdomyosarcoma (ERMS)-a common pediatric cancer of muscle. Here, we used a zebrafish transgenic model of ERMS to identify a role for intracellular NOTCH1 (ICN1) in increasing TPCs by 23-fold. ICN1 expanded TPCs by enabling the de-differentiation of zebrafish ERMS cells into self-renewing myf5+ TPCs, breaking the rigid differentiation hierarchies reported in normal muscle. ICN1 also had conserved roles in regulating human ERMS self-renewal and growth. Mechanistically, ICN1 upregulated expression of SNAIL1, a transcriptional repressor, to increase TPC number in human ERMS and to block muscle differentiation through suppressing MEF2C, a myogenic differentiation transcription factor. Our data implicate the NOTCH1/SNAI1/MEF2C signaling axis as a major determinant of TPC self-renewal and differentiation in ERMS, raising hope of therapeutically targeting this pathway in the future.


Assuntos
Fatores de Transcrição MEF2/metabolismo , Receptor Notch1/metabolismo , Rabdomiossarcoma Embrionário/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Animais , Diferenciação Celular/fisiologia , Humanos , Rabdomiossarcoma Embrionário/patologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas de Xenopus/metabolismo , Peixe-Zebra
12.
Cell Rep ; 18(3): 804-815, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28099856

RESUMO

Zika virus is an emerging arthropod-borne flavivirus for which there are no vaccines or specific therapeutics. We screened a library of 2,000 bioactive compounds for their ability to block Zika virus infection in three distinct cell types with two different strains of Zika virus. Using a microscopy-based assay, we validated 38 drugs that inhibited Zika virus infection, including FDA-approved nucleoside analogs. Cells expressing high levels of the attachment factor AXL can be protected from infection with receptor tyrosine kinase inhibitors, while placental-derived cells that lack AXL expression are insensitive to this inhibition. Importantly, we identified nanchangmycin as a potent inhibitor of Zika virus entry across all cell types tested, including physiologically relevant primary cells. Nanchangmycin also was active against other medically relevant viruses, including West Nile, dengue, and chikungunya viruses that use a similar route of entry. This study provides a resource of small molecules to study Zika virus pathogenesis.


Assuntos
Antivirais/farmacologia , Éteres/química , Compostos de Espiro/química , Zika virus/efeitos dos fármacos , Antivirais/química , Células Cultivadas , Vírus Chikungunya/efeitos dos fármacos , Vírus da Dengue/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/virologia , Éteres/farmacologia , Humanos , Nucleosídeos/química , Nucleosídeos/farmacologia , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Compostos de Espiro/farmacologia , Trofoblastos/citologia , Trofoblastos/virologia , Internalização do Vírus/efeitos dos fármacos , Vírus do Nilo Ocidental/efeitos dos fármacos , Zika virus/fisiologia
13.
PLoS Pathog ; 11(10): e1005168, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26426272

RESUMO

White-nose syndrome (WNS) in North American bats is caused by an invasive cutaneous infection by the psychrophilic fungus Pseudogymnoascus destructans (Pd). We compared transcriptome-wide changes in gene expression using RNA-Seq on wing skin tissue from hibernating little brown myotis (Myotis lucifugus) with WNS to bats without Pd exposure. We found that WNS caused significant changes in gene expression in hibernating bats including pathways involved in inflammation, wound healing, and metabolism. Local acute inflammatory responses were initiated by fungal invasion. Gene expression was increased for inflammatory cytokines, including interleukins (IL) IL-1ß, IL-6, IL-17C, IL-20, IL-23A, IL-24, and G-CSF and chemokines, such as Ccl2 and Ccl20. This pattern of gene expression changes demonstrates that WNS is accompanied by an innate anti-fungal host response similar to that caused by cutaneous Candida albicans infections. However, despite the apparent production of appropriate chemokines, immune cells such as neutrophils and T cells do not appear to be recruited. We observed upregulation of acute inflammatory genes, including prostaglandin G/H synthase 2 (cyclooxygenase-2), that generate eicosanoids and other nociception mediators. We also observed differences in Pd gene expression that suggest host-pathogen interactions that might determine WNS progression. We identified several classes of potential virulence factors that are expressed in Pd during WNS, including secreted proteases that may mediate tissue invasion. These results demonstrate that hibernation does not prevent a local inflammatory response to Pd infection but that recruitment of leukocytes to the site of infection does not occur. The putative virulence factors may provide novel targets for treatment or prevention of WNS. These observations support a dual role for inflammation during WNS; inflammatory responses provide protection but excessive inflammation may contribute to mortality, either by affecting torpor behavior or causing damage upon emergence in the spring.


Assuntos
Quirópteros/genética , Quirópteros/imunologia , Quirópteros/microbiologia , Micoses/veterinária , Animais , Ascomicetos/patogenicidade , Hibernação/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , Micoses/genética , Micoses/imunologia , Síndrome , Transcriptoma , Fatores de Virulência/imunologia , Asas de Animais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...