Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 98(5): e0023924, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38647327

RESUMO

Dengue virus (DENV) represents a significant global health burden, with 50% of the world's population at risk of infection, and there is an urgent need for next-generation vaccines. Virus-like particle (VLP)-based vaccines, which mimic the antigenic structure of the virus but lack the viral genome, are an attractive approach. Here, we describe a dengue VLP (DENVLP) vaccine which generates a neutralizing antibody response against all four DENV serotypes in 100% of immunized non-human primates for up to 1 year. Additionally, DENVLP vaccination produced no ADE response against any of four DENV serotypes in vitro. DENVLP vaccination reduces viral replication in a non-human primate challenge model. We also show that transfer of purified IgG from immunized monkeys into immunodeficient mice protects against subsequent lethal DENV challenge, indicating a humoral mechanism of protection. These results indicate that this DENVLP vaccine is immunogenic and can be considered for clinical evaluation. Immunization of non-human primates with a tetravalent DENVLP vaccine induces high levels of neutralizing antibodies and reduces the severity of infection for all four dengue serotypes.IMPORTANCEDengue is a viral disease that infects nearly 400 million people worldwide and causes dengue hemorrhagic fever, which is responsible for 10,000 deaths each year. Currently, there is no therapeutic drug licensed to treat dengue infection, which makes the development of an effective vaccine essential. Virus-like particles (VLPs) are a safe and highly immunogenic platform that can be used in young children, immunocompromised individuals, as well as healthy adults. In this study, we describe the development of a dengue VLP vaccine and demonstrate that it induces a robust immune response against the dengue virus for over 1 year in monkeys. The immunity induced by this vaccine reduced live dengue infection in both murine and non-human primate models. These results indicate that our dengue VLP vaccine is a promising vaccine candidate.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vacinas contra Dengue , Vírus da Dengue , Dengue , Vacinas de Partículas Semelhantes a Vírus , Animais , Feminino , Camundongos , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Dengue/prevenção & controle , Dengue/imunologia , Dengue/virologia , Vacinas contra Dengue/imunologia , Vacinas contra Dengue/administração & dosagem , Vírus da Dengue/imunologia , Modelos Animais de Doenças , Imunoglobulina G/imunologia , Macaca fascicularis , Macaca mulatta , Sorogrupo , Vacinação , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Replicação Viral
2.
Immunity ; 41(3): 493-502, 2014 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-25238099

RESUMO

The viral accessory protein Vpx, expressed by certain simian and human immunodeficiency viruses (SIVs and HIVs), is thought to improve viral infectivity of myeloid cells. We infected 35 Asian macaques and African green monkeys with viruses that do or do not express Vpx and examined viral targeting of cells in vivo. While lack of Vpx expression affected viral dynamics in vivo, with decreased viral loads and infection of CD4⁺ T cells, Vpx expression had no detectable effect on infectivity of myeloid cells. Moreover, viral DNA was observed only within myeloid cells in tissues not massively depleted of CD4⁺ T cells. Myeloid cells containing viral DNA also showed evidence of T cell phagocytosis in vivo, suggesting that their viral DNA may be attributed to phagocytosis of SIV-infected T cells. These data suggest that myeloid cells are not a major source of SIV in vivo, irrespective of Vpx expression.


Assuntos
Linfócitos T CD4-Positivos/virologia , DNA Viral/análise , Células Mieloides/virologia , Vírus da Imunodeficiência Símia/genética , Proteínas Virais Reguladoras e Acessórias/biossíntese , Animais , Linfócitos T CD4-Positivos/imunologia , Chlorocebus aethiops , Depleção Linfocítica , Macaca , Proteínas Monoméricas de Ligação ao GTP/biossíntese , Fagocitose , Síndrome de Imunodeficiência Adquirida dos Símios , Carga Viral , Proteínas Virais Reguladoras e Acessórias/genética
3.
BMC Musculoskelet Disord ; 24(1): 388, 2023 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-37194069

RESUMO

BACKGROUND: While fear of movement is an important predictor of pain and disability in osteoarthritis (OA), its impact on patients with hip OA remains uncertain. This study aimed to determine whether fear of movement, evaluated by the Tampa Scale for Kinesiophobia (TSK)-11, and pain catastrophizing, evaluated by the Pain Catastrophizing Scale (PCS), were associated with quality of life (QOL) in patients with hip OA. METHODS: This cross-sectional study was conducted between November 2017 and December 2018. Ninety-one consecutively enrolled patients with severe hip OA were scheduled for primary unilateral total hip arthroplasty. The EuroQOL-5 Dimensions questionnaire was used to measure general QOL. The Japanese Orthopedic Association Hip Disease Evaluation Questionnaire was used to assess disease-specific QOL. The covariates included age, sex, body mass index (BMI), pain intensity, high pain catastrophizing (PCS ≥ 30), and high kinesiophobia (TSK-11 ≥ 25). Variables were subjected to multivariate analysis using each QOL scale. RESULTS: In multiple regression analysis, pain intensity, high pain catastrophizing, and BMI were independently correlated with the disease-specific QOL scale. High pain catastrophizing, pain intensity, and high kinesiophobia were independently correlated with the general QOL scale. CONCLUSIONS: High pain catastrophizing (PCS ≥ 30) was independently associated with disease and general QOL scales. High kinesiophobia (TSK-11 ≥ 25) was independently associated with the general QOL scale in preoperative patients with severe hip OA.


Assuntos
Osteoartrite do Quadril , Humanos , Osteoartrite do Quadril/complicações , Osteoartrite do Quadril/cirurgia , Cinesiofobia , Estudos Transversais , Qualidade de Vida , Catastrofização , Dor
4.
BMC Musculoskelet Disord ; 22(1): 482, 2021 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-34034743

RESUMO

BACKGROUND: Hip arthroplasty using acrylic prosthesis was once conducted; however, it has now been abandoned because of early breakages and wear of material. Therefore, complications or revision surgeries due to the use of acrylic prostheses are becoming rare. CASE PRESENTATION: A 76-year-old woman presented with a sudden onset of severe pain in her left femur while walking. Radiographs revealed severe osteolysis and periprosthetic femoral fracture 45 years after the initial hemiarthroplasty using an acrylic prosthesis. We performed a Girdlestone resection arthroplasty by removing the prosthesis and fixing the fracture site using an intramedullary nail and metal plate. The patient was pain-free in her hip and leg 2 years and 9 months after the surgery. Although she walked with a cane or crutches, no postoperative complications were observed. CONCLUSIONS: The combined use of an intramedullary nail and plate fixation with resection arthroplasty could offer acceptable results for patients with severe osteolysis and periprosthetic fracture after femoral head replacement using an acrylic prosthesis. Our report seems relevant because it not only reminds us of the significant steps made in the development of modern total hip arthroplasty; it also highlights one of the surgical options for severe osteolysis and periprosthetic fracture of the hip.


Assuntos
Artroplastia de Quadril , Fraturas do Fêmur , Hemiartroplastia , Prótese de Quadril , Osteólise , Fraturas Periprotéticas , Idoso , Artroplastia de Quadril/efeitos adversos , Feminino , Fraturas do Fêmur/cirurgia , Fêmur/cirurgia , Hemiartroplastia/efeitos adversos , Prótese de Quadril/efeitos adversos , Humanos , Osteólise/diagnóstico por imagem , Osteólise/etiologia , Osteólise/cirurgia , Fraturas Periprotéticas/diagnóstico por imagem , Fraturas Periprotéticas/etiologia , Fraturas Periprotéticas/cirurgia , Reoperação
5.
PLoS Pathog ; 13(8): e1006538, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28787449

RESUMO

Despite effective control of plasma viremia with the use of combination antiretroviral therapies (cART), minor cognitive and motor disorders (MCMD) persist as a significant clinical problem in HIV-infected patients. Non-human primate models are therefore required to study mechanisms of disease progression in the central nervous system (CNS). We isolated a strain of simian immunodeficiency virus (SIV), SIVsm804E, which induces neuroAIDS in a high proportion of rhesus macaques and identified enhanced antagonism of the host innate factor BST-2 as an important factor in the macrophage tropism and initial neuro-invasion of this isolate. In the present study, we further developed this model by deriving a molecular clone SIVsm804E-CL757 (CL757). This clone induced neurological disorders in high frequencies but without rapid disease progression and thus is more reflective of the tempo of neuroAIDS in HIV-infection. NeuroAIDS was also induced in macaques co-inoculated with CL757 and the parental AIDS-inducing, but non-neurovirulent SIVsmE543-3 (E543-3). Molecular analysis of macaques infected with CL757 revealed compartmentalization of virus populations between the CNS and the periphery. CL757 exclusively targeted the CNS whereas E543-3 was restricted to the periphery consistent with a role for viral determinants in the mechanisms of neuroinvasion. CL757 would be a useful model to investigate disease progression in the CNS and as a model to study virus reservoirs in the CNS.


Assuntos
Complexo AIDS Demência/virologia , Modelos Animais de Doenças , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Vírus da Imunodeficiência Símia/genética , Animais , Encéfalo/virologia , Citometria de Fluxo , Macaca mulatta , Reação em Cadeia da Polimerase , Síndrome de Imunodeficiência Adquirida dos Símios/complicações
6.
J Virol ; 90(9): 4520-4529, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26912615

RESUMO

UNLABELLED: Mucosa-associated invariant T (MAIT) cells contribute to host immune protection against a wide range of potential pathogens via the recognition of bacterial metabolites presented by the major histocompatibility complex class I-related molecule MR1. Although bacterial products translocate systemically in human immunodeficiency virus (HIV)-infected individuals and simian immunodeficiency virus (SIV)-infected Asian macaques, several studies have shown that MAIT cell frequencies actually decrease in peripheral blood during the course of HIV/SIV disease. However, the mechanisms underlying this proportional decline remain unclear. In this study, we characterized the phenotype, activation status, functionality, distribution, and clonotypic structure of MAIT cell populations in the peripheral blood, liver, mesenteric lymph nodes (MLNs), jejunum, and bronchoalveolar lavage (BAL) fluid of healthy and SIV-infected rhesus macaques (RMs). Low frequencies of MAIT cells were observed in the peripheral blood, MLNs, and BAL fluid of SIV-infected RMs. These numerical changes were coupled with increased proliferation and a highly public T cell receptor alpha (TCRα) repertoire in the MAIT cell compartment without redistribution to other anatomical sites. Collectively, our data show systemically decreased frequencies of MAIT cells likely attributable to enhanced turnover in SIV-infected RMs. This process may impair protective immunity against certain opportunistic infections with progression to AIDS. IMPORTANCE: The data presented in this study reveal for the first time that MAIT cells are systemically depleted in an AIDS virus infection. These findings provide a new mechanistic link with our current understanding of HIV/SIV pathogenesis and implicate MAIT cell depletion in the disease process.


Assuntos
Mucosa/citologia , Mucosa/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Subpopulações de Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Movimento Celular/imunologia , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/genética , Imunidade nas Mucosas , Imunomodulação , Imunofenotipagem , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Depleção Linfocítica , Macaca mulatta , Mucosa/metabolismo , Fragmentos de Peptídeos/química , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Fenótipo , Matrizes de Pontuação de Posição Específica , Receptores de Antígenos de Linfócitos T alfa-beta/química , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Subpopulações de Linfócitos T/metabolismo , Carga Viral
7.
J Virol ; 90(24): 11087-11095, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27681142

RESUMO

TRIM5α polymorphism limits and complicates the use of simian immunodeficiency virus (SIV) for evaluation of human immunodeficiency virus (HIV) vaccine strategies in rhesus macaques. We previously reported that the TRIM5α-sensitive SIV from sooty mangabeys (SIVsm) clone SIVsmE543-3 acquired amino acid substitutions in the capsid that overcame TRIM5α restriction when it was passaged in rhesus macaques expressing restrictive TRIM5α alleles. Here we generated TRIM5α-resistant clones of the related SIVsmE660 strain without animal passage by introducing the same amino acid capsid substitutions. We evaluated one of the variants in rhesus macaques expressing permissive and restrictive TRIM5α alleles. The SIVsmE660 variant infected and replicated in macaques with restrictive TRIM5α genotypes as efficiently as in macaques with permissive TRIM5α genotypes. These results demonstrated that mutations in the SIV capsid can confer SIV resistance to TRIM5α restriction without animal passage, suggesting an applicable method to generate more diverse SIV strains for HIV vaccine studies. IMPORTANCE: Many strains of SIV from sooty mangabey monkeys are susceptible to resistance by common rhesus macaque TRIM5α alleles and result in reduced virus acquisition and replication in macaques that express these restrictive alleles. We previously observed that spontaneous variations in the capsid gene were associated with improved replication in macaques, and the introduction of two amino acid changes in the capsid transfers this improved replication to the parent clone. In the present study, we introduced these mutations into a related but distinct strain of SIV that is commonly used for challenge studies for vaccine trials. These mutations also improved the replication of this strain in macaques with the restrictive TRIM5α genotype and thus will eliminate the confounding effects of TRIM5α in vaccine studies.


Assuntos
Capsídeo/imunologia , Proteínas de Transporte/genética , Evasão da Resposta Imune , RNA Viral/genética , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/imunologia , Alelos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Capsídeo/química , Proteínas de Transporte/imunologia , Cercocebus atys , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Mutação , RNA Viral/imunologia , Alinhamento de Sequência , Transdução de Sinais , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/mortalidade , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/patogenicidade , Análise de Sobrevida , Dedos de Zinco
8.
J Virol ; 89(4): 2233-40, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25473059

RESUMO

UNLABELLED: Tripartite motif-containing protein 5α (TRIM5α) is considered to be a potential target for cell-based gene modification therapy against human immunodeficiency virus type 1 (HIV-1) infection. In the present study, we used a relevant rhesus macaque model of infection with simian immunodeficiency virus from sooty mangabey (SIVsm) to evaluate the effect of TRIM5α restriction on clinical outcome. For macaques expressing a restrictive TRIM5 genotype, the disease outcomes of those infected with the wild-type TRIM-sensitive SIVsm strain and those infected with a virus with escape mutations in the capsid were compared. We found that TRIM5α restriction significantly delayed disease progression and improved the survival rate of SIV-infected macaques, supporting the feasibility of exploiting TRIM5α as a target for gene therapy against HIV-1. Furthermore, we also found that preservation of memory CD4 T cells was associated with protection by TRIM5α restriction, suggesting memory CD4 T cells or their progenitor cells as an ideal target for gene modification. Despite the significant effect of TRIM5α restriction on survival, SIV escape from TRIM5α restriction was also observed; therefore, this may not be an effective stand-alone strategy and may require combination with other targets. IMPORTANCE: Recent studies suggest that it may be feasible not only to suppress viral replication with antiviral drugs but also potentially to eliminate or "cure" human immunodeficiency virus (HIV) infection. One approach being explored is the use of gene therapy to introduce genes that can restrict HIV replication, including a restrictive version of the host factor TRIM5α. TRIM5 was identified as a factor that restricts HIV replication in macaque cells. The rhesus gene is polymorphic, and some alleles are restrictive for primary SIVsm isolates, although escape mutations arise late in infection. Introduction of these escape mutations into the parental virus conferred resistance to TRIM5 on macaques. The present study evaluated these animals for long-term outcomes and found that TRIM5α restriction significantly delayed disease progression and improved the survival rate of SIV-infected macaques, suggesting that this could be a valid gene therapy approach that could be adapted for HIV.


Assuntos
Proteínas/imunologia , Proteínas/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Progressão da Doença , Genótipo , Memória Imunológica , Macaca mulatta , Proteínas/genética , Síndrome de Imunodeficiência Adquirida dos Símios/mortalidade , Análise de Sobrevida , Ubiquitina-Proteína Ligases
9.
J Virol ; 90(5): 2316-31, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26656714

RESUMO

UNLABELLED: African green monkeys (AGM) are natural hosts of simian immunodeficiency virus (SIV), and infection in these animals is generally nonpathogenic, whereas infection of nonnatural hosts, such as rhesus macaques (RM), is commonly pathogenic. CCR5 has been described as the primary entry coreceptor for SIV in vivo, while human-derived CXCR6 and GPR15 also appear to be used in vitro. However, sooty mangabeys that are genetically deficient in CCR5 due to an out-of-frame deletion are infectible with SIVsmm, indicating that SIVsmm can use alternative coreceptors in vivo. In this study, we examined the CCR5 dependence of SIV strains derived from vervet AGM (SIVagmVer) and the ability of AGM-derived GPR15 and CXCR6 to serve as potential entry coreceptors. We found that SIVagmVer replicated efficiently in AGM and RM peripheral blood mononuclear cells (PBMC) in the presence of the CCR5 antagonist maraviroc, despite the fact that maraviroc was capable of blocking the CCR5-tropic strains SIVmac239, SIVsmE543-3, and simian-human immunodeficiency virus SHIV-AD8 in RM PBMC. We also found that AGM CXCR6 and AGM GPR15, to a lesser extent, supported entry of pseudotype viruses bearing SIVagm envelopes, including SIVagm transmitted/founder envelopes. Lastly, we found that CCR5, GPR15, and CXCR6 mRNAs were detected in AGM and RM memory CD4(+) T cells. These results suggest that GPR15 and CXCR6 are expressed on AGM CD4(+) T cells and are potential alternative coreceptors for SIVagm use in vivo. These data suggest that the use of non-CCR5 entry pathways may be a common feature of SIV replication in natural host species, with the potential to contribute to nonpathogenicity in these animals. IMPORTANCE: African green monkeys (AGM) are natural hosts of SIV, and infection in these animals generally does not cause AIDS, whereas SIV-infected rhesus macaques (RM) typically develop AIDS. Although it has been reported that SIV generally uses CD4 and CCR5 to enter target cells in vivo, other molecules, such as GPR15 and CXCR6, also function as SIV coreceptors in vitro. In this study, we investigated whether SIV from vervet AGM can use non-CCR5 entry pathways, as has been observed in sooty mangabeys. We found that SIVagmVer efficiently replicated in AGM and RM peripheral blood mononuclear cells in the presence of the CCR5 antagonist maraviroc, suggesting that non-CCR5 entry pathways can support SIVagm entry. We found that AGM-derived GPR15 and CXCR6 support SIVagmVer entry in vitro and may serve as entry coreceptors for SIVagm in vivo, since their mRNAs were detected in AGM memory CD4(+) T cells, the preferred target cells of SIV.


Assuntos
Linfócitos/virologia , Receptores de Quimiocinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virais/metabolismo , Vírus da Imunodeficiência Símia/fisiologia , Internalização do Vírus , Animais , Células Cultivadas , Chlorocebus aethiops , Replicação Viral
10.
J Gen Virol ; 96(9): 2867-2877, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26041873

RESUMO

It has been estimated that human immunodeficiency virus type 1 originated from the zoonotic transmission of simian immunodeficiency virus (SIV) of chimpanzees, SIVcpz, and that SIVcpz emerged by the recombination of two lineages of SIVs in Old World monkeys (SIVgsn/mon/mus in guenons and SIVrcm in red-capped mangabeys) and SIVcpz Nef is most closely related to SIVrcm Nef. These observations suggest that SIVrcm Nef had an advantage over SIVgsn/mon/mus during the evolution of SIVcpz in chimpanzees, although this advantage remains uncertain. Nef is a multifunctional protein which downregulates CD4 and coreceptor proteins from the surface of infected cells, presumably to limit superinfection. To assess the possibility that SIVrcm Nef was selected by its superior ability to downregulate viral entry receptors in chimpanzees, we compared its ability to down-modulate viral receptor proteins from humans, chimpanzees and red-capped mangabeys with Nef proteins from eight other different strains of SIVs. Surprisingly, the ability of SIVrcm Nef to downregulate CCR5, CCR2B and CXCR6 was comparable to or lower than SIVgsn/mon/mus Nef, indicating that ability to down-modulate chemokine receptors was not the selective pressure. However, SIVrcm Nef significantly downregulates chimpanzee CD4 over SIVgsn/mon/mus Nefs. Our findings suggest the possibility that the selection of SIVrcm Nef by ancestral SIVcpz is due to its superior capacity to down-modulate chimpanzees CD4 rather than coreceptor proteins.


Assuntos
Evolução Molecular , Produtos do Gene nef/genética , Lentivirus de Primatas/genética , Doenças dos Primatas/genética , Receptores Virais/genética , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Vírus da Imunodeficiência Símia/genética , Animais , Cercocebus , Produtos do Gene nef/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Lentivirus de Primatas/classificação , Lentivirus de Primatas/metabolismo , Pan troglodytes , Filogenia , Doenças dos Primatas/metabolismo , Doenças dos Primatas/virologia , Primatas , Receptores Virais/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/classificação , Vírus da Imunodeficiência Símia/metabolismo
11.
J Virol ; 88(22): 13201-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25187546

RESUMO

UNLABELLED: Although nonhuman primate models of neuro-AIDS have made tremendous contributions to our understanding of disease progression in the central nervous system (CNS) of human immunodeficiency virus type 1 (HIV-1)-infected individuals, each model holds advantages and limitations. In this study, in vivo passage of SIVsmE543 was conducted to obtain a viral isolate that can induce neuropathology in rhesus macaques. After a series of four in vivo passages in rhesus macaques, we have successfully isolated SIVsm804E. SIVsm804E shows efficient replication in peripheral blood mononuclear cells (PBMCs) and monocyte-derived macrophages (MDMs) in vitro and induces neuro-AIDS in high frequencies in vivo. Analysis of the acute phase of infection revealed that SIVsm804E establishes infection in the CNS during the early phase of the infection, which was not observed in the animals infected with the parental SIVsmE543-3. Comprehensive analysis of disease progression in the animals used in the study suggested that host major histocompatibility complex class I (MHC-I) and TRIM5α genotypes influence the disease progression in the CNS. Taken together, our findings show that we have successfully isolated a new strain of simian immunodeficiency virus (SIV) that is capable of establishing infection in the CNS at early stage of infection and causes neuropathology in infected rhesus macaques at a high frequency (83%) using a single inoculum, when animals with restrictive MHC-I or TRIM5α genotypes are excluded. SIVsm804E has the potential to augment some of the limitations of existing nonhuman primate neuro-AIDS models. IMPORTANCE: Human immunodeficiency virus (HIV) is associated with a high frequency of neurologic complications due to infection of the central nervous system (CNS). Although the use of antiviral treatment has reduced the incidence of severe complications, milder disease of the CNS continues to be a significant problem. Animal models to study development of neurologic disease are needed. This article describes the development of a novel virus isolate that induces neurologic disease in a high proportion of rhesus macaques infected without the need for prior immunomodulation as is required for some other models.


Assuntos
Encefalite Viral/imunologia , Macaca mulatta , Complexo Principal de Histocompatibilidade/imunologia , Proteínas/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Animais , Encéfalo/virologia , Encefalite Viral/genética , Genótipo , Lentivirus de Primatas , Complexo Principal de Histocompatibilidade/genética , Dados de Sequência Molecular , Proteínas/genética , Análise de Sequência de DNA , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Ubiquitina-Proteína Ligases , Virulência
12.
PLoS Pathog ; 9(8): e1003577, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23990789

RESUMO

The antagonistic interaction with host restriction proteins is a major driver of evolutionary change for viruses. We previously reported that polymorphisms of the TRIM5α B30.2/SPRY domain impacted the level of SIVsmm viremia in rhesus macaques. Viremia in macaques homozygous for the non-restrictive TRIM5α allele TRIM5(Q) was significantly higher than in macaques expressing two restrictive TRIM5alpha alleles TRIM5(TFP/TFP) or TRIM5(Cyp/TFP). Using this model, we observed that despite an early impact on viremia, SIVsmm overcame TRIM5α restriction at later stages of infection and that increasing viremia was associated with specific amino acid substitutions in capsid. Two amino acid substitutions (P37S and R98S) in the capsid region were associated with escape from TRIM5(TFP) restriction and substitutions in the CypA binding-loop (GPLPA87-91) in capsid were associated with escape from TRIM5(Cyp). Introduction of these mutations into the original SIVsmE543 clone not only resulted in escape from TRIM5α restriction in vitro but the P37S and R98S substitutions improved virus fitness in macaques with homozygous restrictive TRIM(TFP) alleles in vivo. Similar substitutions were observed in other SIVsmm strains following transmission and passage in macaques, collectively providing direct evidence that TRIM5α exerts selective pressure on the cross-species transmission of SIV in primates.


Assuntos
Alelos , Proteínas de Transporte/metabolismo , Evolução Molecular , Mutação de Sentido Incorreto , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Vírus da Imunodeficiência Símia/metabolismo , Substituição de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/genética , Macaca mulatta , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão , Vírus da Imunodeficiência Símia/genética , Viremia/genética , Viremia/metabolismo
13.
J Virol ; 87(16): 8896-908, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23720733

RESUMO

Nonhuman primate-simian immunodeficiency virus (SIV) models are powerful tools for studying the pathogenesis of human immunodeficiency virus type 1 (HIV-1) in the brain. Our laboratory recently isolated a neuropathogenic viral swarm, SIVsmH804E, a derivative of SIVsmE543-3, which was the result of sequential intravenous passages of viruses isolated from the brains of rhesus macaques with SIV encephalitis. Animals infected with SIVsmH804E or its precursor (SIVsmH783Br) developed SIV meningitis and/or encephalitis at high frequencies. Since we observed macaques with a combination of meningitis and encephalitis, as well as animals in which meningitis or encephalitis was the dominant component, we hypothesized that distinct mechanisms could be driving the two pathological states. Therefore, we assessed viral populations in the meninges and the brain parenchyma by laser capture microdissection. Viral RNAs were isolated from representative areas of the meninges, brain parenchyma, terminal plasma, and cerebrospinal fluid (CSF) and from the inoculum, and the SIV envelope fragment was amplified by PCR. Phylogenetic analysis of envelope sequences from the conventional progressors revealed compartmentalization of viral populations between the meninges and the parenchyma. In one of these animals, viral populations in meninges were closely related to those from CSF and shared signature truncations in the cytoplasmic domain of gp41, consistent with a common origin. Apart from magnetic resonance imaging (MRI) and positron-emission tomography (PET) imaging, CSF is the most accessible assess to the central nervous system for HIV-1-infected patients. However, our results suggest that the virus in the CSF may not always be representative of viral populations in the brain and that caution should be applied in extrapolating between the properties of viruses in these two compartments.


Assuntos
Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Encefalite Viral/patologia , Meningite Viral/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/patogenicidade , Animais , Líquido Cefalorraquidiano/virologia , Análise por Conglomerados , Encefalite Viral/virologia , Produtos do Gene env/genética , Microdissecção e Captura a Laser , Macaca mulatta , Meninges/virologia , Meningite Viral/virologia , Dados de Sequência Molecular , Filogenia , Plasma/virologia , Reação em Cadeia da Polimerase , RNA Viral/genética , RNA Viral/isolamento & purificação , Análise de Sequência de DNA , Homologia de Sequência , Vírus da Imunodeficiência Símia/isolamento & purificação , Virulência
14.
Arthroplasty ; 6(1): 22, 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38704579

RESUMO

BACKGROUND: Implant impingement and soft tissue tension are factors involved in dislocation after total hip arthroplasty (THA). Combined anteversion (CA) has been used as an indicator for implant placement. However, optimal implant placement remains a challenge. Moreover, the effect of changes in offset on dislocation is still unclear. In this study, we aimed to clarify the effects of postoperative CA and pre- and postoperative changes in offset on dislocation. METHODS: Included were patients who underwent primary cementless THA between 2013 and 2020. The mean values of CA and offset in the dislocation and non-dislocation groups were compared. The CA values within ± 10% of the recommended values were defined as good CA, and those outside the range were rated as poor CA. The dislocation rates were compared between the good and poor CA groups and between the groups with and without increased offset. RESULTS: A total of 283 hips were included. The mean values of CA in the dislocation and non-dislocation groups were significantly different (P < 0.05). The dislocation rate was significantly lower in the good CA group (P < 0.05). The dislocation rates in the groups with and without increased total offset were 0.5% and 4.3%, respectively (P = 0.004). There were no dislocations in patients with good CA and increased offset. CONCLUSIONS: The dislocation rate was significantly lower when implants were placed within ± 10% of the recommended CA value. Our results suggest that dislocation can be avoided by placing the implant in the good CA range and considering the increase in total offset on the operative side.

15.
iScience ; 27(2): 108964, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38352232

RESUMO

Continuing emergence of variants of concern resulting in reduced SARS-CoV-2 vaccine efficacy necessitates additional prevention strategies. The structure of VLPCOV-01, a lipid nanoparticle-encapsulated, self-amplifying RNA COVID-19 vaccine with a comparable immune response to BNT162b2, was revised by incorporating a modified base, 5-methylcytosine, to reduce reactogenicity, and an updated receptor-binding domain derived from the Brazil (gamma) variant. Interim analyses of a phase 1 dose-escalation booster vaccination study with the resulting construct, VLPCOV-02, in healthy, previously vaccinated Japanese individuals (N = 96) are reported (jRCT2051230005). A dose-related increase in solicited local and systemic adverse events was observed, which were generally rated mild or moderate. The most commonly occurring events were tenderness, pain, fatigue, and myalgia. Serum SARS-CoV-2 immunoglobulin titers increased during the 4 weeks post-immunization. VLPCOV-02 demonstrated a favorable safety profile compared with VLPCOV-01, with reduced adverse events and fewer fever events at an equivalent dose. These findings support further study of VLPCOV-02.

16.
J Virol ; 86(24): 13795-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23035225

RESUMO

Simian immunodeficiency virus (SIV) infection of macaques can result in central nervous system disorders, such as meningitis and encephalitis. We studied 10 animals inoculated with brain-derived virus from animals with SIV encephalitis. Over half of the macaques developed SIV-induced neurologic disease. Elevated levels of systemic immune activation were observed to correlate with viral RNA in the cerebral spinal fluid but not with plasma viral load, consistent with a role for SIV in the pathogenesis of neurologic disease.


Assuntos
Doenças do Sistema Nervoso/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Animais , Macaca , Doenças do Sistema Nervoso/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia
17.
Int J Med Robot ; : e2613, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38108101

RESUMO

BACKGROUND: The aim of this study is to determine whether the initial stability of a cementless cup with the Mako system is superior to that of a conventional manual technique using bone models. METHODS: The bone models were prepared using a polyurethane foam block. Two hemispherical cementless cups (highly porous titanium cup [Trident II Tritanium, Stryker] and hydroxyapatite-coated titanium cup [Trident HA, Stryker]) were implanted using the Mako system. The torque of the cups was measured by rotational and lever-out torque testing and compared with that of a conventional manual technique. RESULTS: The two types of cups that were implanted using the Mako system demonstrated significantly higher mean rotational torque than that of the manual technique (p < 0.01, p = 0.01, respectively). CONCLUSIONS: This study provides the advantage of the initial stability of a cementless hemispherical cup implanted by the Mako system compared with that of the conventional manual technique.

18.
bioRxiv ; 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37961509

RESUMO

In order to improve vaccine effectiveness and safety profile of existing synthetic RNA-based vaccines, we have developed a self-amplifying RNA (saRNA)-based vaccine expressing membrane-anchored receptor binding domain (RBD) of SARS-CoV-2 S protein (S-RBD) and have demonstrated that a minimal dose of this saRNA vaccine elicits robust immune responses. Results from a recent clinical trial with 5-methylcytidine (5mC) incorporating saRNA vaccine demonstrated reduced vaccine-induced adverse effects while maintaining robust humoral responses. In this study, we investigate the mechanisms accounting for induction of efficient innate and adaptive immune responses and attenuated adverse effects induced by the 5mC-incorporated saRNA. We show that the 5mC-incorporating saRNA platform leads to prolonged and robust expression of antigen, while induction of type-I interferon (IFN-I), a key driver of reactogenicity, is attenuated in peripheral blood mononuclear cells (PBMCs), but not in macrophages and dendritic cells. Interestingly, we find that the major cellular source of IFN-I production in PBMCs is plasmacytoid dendritic cells (pDCs), which is attenuated upon 5mC incorporation in saRNA. In addition, we demonstrate that monocytes also play an important role in amplifying proinflammatory responses. Furthermore, we show that the detection of saRNA is mediated by a host cytosolic RNA sensor, RIG-I. Importantly, 5mC-incorporating saRNA vaccine candidate produced robust IgG responses against S-RBD upon injection in mice, thus providing strong support for the potential clinical use of 5mC-incorporating saRNA vaccines.

19.
Nat Commun ; 14(1): 2810, 2023 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-37208330

RESUMO

Several vaccines have been widely used to counteract the global pandemic caused by SARS-CoV-2. However, due to the rapid emergence of SARS-CoV-2 variants of concern (VOCs), further development of vaccines that confer broad and longer-lasting protection against emerging VOCs are needed. Here, we report the immunological characteristics of a self-amplifying RNA (saRNA) vaccine expressing the SARS-CoV-2 Spike (S) receptor binding domain (RBD), which is membrane-anchored by fusing with an N-terminal signal sequence and a C-terminal transmembrane domain (RBD-TM). Immunization with saRNA RBD-TM delivered in lipid nanoparticles (LNP) efficiently induces T-cell and B-cell responses in non-human primates (NHPs). In addition, immunized hamsters and NHPs are protected against SARS-CoV-2 challenge. Importantly, RBD-specific antibodies against VOCs are maintained for at least 12 months in NHPs. These findings suggest that this saRNA platform expressing RBD-TM will be a useful vaccine candidate inducing durable immunity against emerging SARS-CoV-2 strains.


Assuntos
COVID-19 , Vacinas , Animais , Cricetinae , Humanos , SARS-CoV-2/genética , COVID-19/prevenção & controle , Motivo de Reconhecimento de RNA , Glicoproteína da Espícula de Coronavírus/genética , Anticorpos Neutralizantes , Anticorpos Antivirais
20.
Cell Rep Med ; 4(8): 101134, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37586325

RESUMO

VLPCOV-01 is a lipid nanoparticle-encapsulated self-amplifying RNA (saRNA) vaccine that expresses a membrane-anchored receptor-binding domain (RBD) derived from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. A phase 1 study of VLPCOV-01 is conducted (jRCT2051210164). Participants who completed two doses of the BNT162b2 mRNA vaccine previously are randomized to receive one intramuscular vaccination of 0.3, 1.0, or 3.0 µg VLPCOV-01, 30 µg BNT162b2, or placebo. No serious adverse events have been reported. VLPCOV-01 induces robust immunoglobulin G (IgG) titers against the RBD protein that are maintained up to 26 weeks in non-elderly participants, with geometric means ranging from 5,037 (95% confidence interval [CI] 1,272-19,940) at 0.3 µg to 12,873 (95% CI 937-17,686) at 3 µg compared with 3,166 (95% CI 1,619-6,191) with 30 µg BNT162b2. Neutralizing antibody titers against all variants of SARS-CoV-2 tested are induced. VLPCOV-01 is immunogenic following low-dose administration. These findings support the potential for saRNA as a vaccine platform.


Assuntos
COVID-19 , Vacinas , Humanos , Pessoa de Meia-Idade , Vacinas contra COVID-19/efeitos adversos , Vacina BNT162 , SARS-CoV-2/genética , RNA , COVID-19/prevenção & controle , Vacinas de mRNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA