Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Cell ; 185(7): 1157-1171.e22, 2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-35259335

RESUMO

Enterococci are a part of human microbiota and a leading cause of multidrug resistant infections. Here, we identify a family of Enterococcus pore-forming toxins (Epxs) in E. faecalis, E. faecium, and E. hirae strains isolated across the globe. Structural studies reveal that Epxs form a branch of ß-barrel pore-forming toxins with a ß-barrel protrusion (designated the top domain) sitting atop the cap domain. Through a genome-wide CRISPR-Cas9 screen, we identify human leukocyte antigen class I (HLA-I) complex as a receptor for two members (Epx2 and Epx3), which preferentially recognize human HLA-I and homologous MHC-I of equine, bovine, and porcine, but not murine, origin. Interferon exposure, which stimulates MHC-I expression, sensitizes human cells and intestinal organoids to Epx2 and Epx3 toxicity. Co-culture with Epx2-harboring E. faecium damages human peripheral blood mononuclear cells and intestinal organoids, and this toxicity is neutralized by an Epx2 antibody, demonstrating the toxin-mediated virulence of Epx-carrying Enterococcus.


Assuntos
Toxinas Bacterianas/metabolismo , Enterococcus , Leucócitos Mononucleares , Fatores de Virulência/metabolismo , Animais , Bovinos , Enterococcus/metabolismo , Enterococcus/patogenicidade , Cavalos , Camundongos , Testes de Sensibilidade Microbiana , Suínos
2.
Cell ; 184(10): 2605-2617.e18, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33831372

RESUMO

Many individuals mount nearly identical antibody responses to SARS-CoV-2. To gain insight into how the viral spike (S) protein receptor-binding domain (RBD) might evolve in response to common antibody responses, we studied mutations occurring during virus evolution in a persistently infected immunocompromised individual. We use antibody Fab/RBD structures to predict, and pseudotypes to confirm, that mutations found in late-stage evolved S variants confer resistance to a common class of SARS-CoV-2 neutralizing antibodies we isolated from a healthy COVID-19 convalescent donor. Resistance extends to the polyclonal serum immunoglobulins of four out of four healthy convalescent donors we tested and to monoclonal antibodies in clinical use. We further show that affinity maturation is unimportant for wild-type virus neutralization but is critical to neutralization breadth. Because the mutations we studied foreshadowed emerging variants that are now circulating across the globe, our results have implications to the long-term efficacy of S-directed countermeasures.


Assuntos
Anticorpos Antivirais/imunologia , COVID-19 , Evolução Molecular , Evasão da Resposta Imune/imunologia , Hospedeiro Imunocomprometido , Fragmentos Fab das Imunoglobulinas/imunologia , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Anticorpos Neutralizantes , COVID-19/genética , COVID-19/imunologia , Feminino , Células HEK293 , Humanos , Masculino , Domínios Proteicos , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia
3.
Mol Cell ; 81(11): 2261-2265, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34087174

RESUMO

COVID-19 altered our lives and pushed scientific research to operate at breakneck speed, leading to significant breakthroughs in record time. We asked experts in the field about the challenges they faced in transitioning, rapidly but safely, to working on the virus while navigating the shutdown. Their voices converge on the importance of teamwork, forging new collaborations, and working toward a shared goal.


Assuntos
Pesquisa Biomédica , COVID-19/epidemiologia , COVID-19/prevenção & controle , Pandemias , Quarentena , SARS-CoV-2 , Humanos , Poesia como Assunto
4.
Nature ; 602(7897): 475-480, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34929721

RESUMO

Alphaviruses, like many other arthropod-borne viruses, infect vertebrate species and insect vectors separated by hundreds of millions of years of evolutionary history. Entry into evolutionarily divergent host cells can be accomplished by recognition of different cellular receptors in different species, or by binding to receptors that are highly conserved across species. Although multiple alphavirus receptors have been described1-3, most are not shared among vertebrate and invertebrate hosts. Here we identify the very low-density lipoprotein receptor (VLDLR) as a receptor for the prototypic alphavirus Semliki forest virus. We show that the E2 and E1 glycoproteins (E2-E1) of Semliki forest virus, eastern equine encephalitis virus and Sindbis virus interact with the ligand-binding domains (LBDs) of VLDLR and apolipoprotein E receptor 2 (ApoER2), two closely related receptors. Ectopic expression of either protein facilitates cellular attachment, and internalization of virus-like particles, a VLDLR LBD-Fc fusion protein or a ligand-binding antagonist block Semliki forest virus E2-E1-mediated infection of human and mouse neurons in culture. The administration of a VLDLR LBD-Fc fusion protein has protective activity against rapidly fatal Semliki forest virus infection in mouse neonates. We further show that invertebrate receptor orthologues from mosquitoes and worms can serve as functional alphavirus receptors. We propose that the ability of some alphaviruses to infect a wide range of hosts is a result of their engagement of evolutionarily conserved lipoprotein receptors and contributes to their pathogenesis.


Assuntos
Mosquitos Vetores , Vírus da Floresta de Semliki , Animais , Proteínas Relacionadas a Receptor de LDL , Ligantes , Camundongos , Receptores de LDL , Vírus da Floresta de Semliki/metabolismo , Sindbis virus/fisiologia
5.
Nature ; 606(7914): 576-584, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35385861

RESUMO

SARS-CoV-2 can cause acute respiratory distress and death in some patients1. Although severe COVID-19 is linked to substantial inflammation, how SARS-CoV-2 triggers inflammation is not clear2. Monocytes and macrophages are sentinel cells that sense invasive infection to form inflammasomes that activate caspase-1 and gasdermin D, leading to inflammatory death (pyroptosis) and the release of potent inflammatory mediators3. Here we show that about 6% of blood monocytes of patients with COVID-19 are infected with SARS-CoV-2. Monocyte infection depends on the uptake of antibody-opsonized virus by Fcγ receptors. The plasma of vaccine recipients does not promote antibody-dependent monocyte infection. SARS-CoV-2 begins to replicate in monocytes, but infection is aborted, and infectious virus is not detected in the supernatants of cultures of infected monocytes. Instead, infected cells undergo pyroptosis mediated by activation of NLRP3 and AIM2 inflammasomes, caspase-1 and gasdermin D. Moreover, tissue-resident macrophages, but not infected epithelial and endothelial cells, from lung autopsies from patients with COVID-19 have activated inflammasomes. Taken together, these findings suggest that antibody-mediated SARS-CoV-2 uptake by monocytes and macrophages triggers inflammatory cell death that aborts the production of infectious virus but causes systemic inflammation that contributes to COVID-19 pathogenesis.


Assuntos
COVID-19 , Inflamação , Monócitos , Receptores de IgG , SARS-CoV-2 , COVID-19/virologia , Caspase 1/metabolismo , Proteínas de Ligação a DNA , Humanos , Inflamassomos/metabolismo , Inflamação/metabolismo , Inflamação/virologia , Monócitos/metabolismo , Monócitos/virologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas de Ligação a Fosfato , Proteínas Citotóxicas Formadoras de Poros , Receptores de IgG/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33879614

RESUMO

The de novo design of polar protein-protein interactions is challenging because of the thermodynamic cost of stripping water away from the polar groups. Here, we describe a general approach for designing proteins which complement exposed polar backbone groups at the edge of beta sheets with geometrically matched beta strands. We used this approach to computationally design small proteins that bind to an exposed beta sheet on the human transferrin receptor (hTfR), which shuttles interacting proteins across the blood-brain barrier (BBB), opening up avenues for drug delivery into the brain. We describe a design which binds hTfR with a 20 nM Kd, is hyperstable, and crosses an in vitro microfluidic organ-on-a-chip model of the human BBB. Our design approach provides a general strategy for creating binders to protein targets with exposed surface beta edge strands.


Assuntos
Engenharia de Proteínas/métodos , Receptores da Transferrina/metabolismo , Receptores da Transferrina/fisiologia , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Sistemas de Liberação de Medicamentos , Humanos , Proteínas/metabolismo , Transferrina/metabolismo
7.
PLoS Pathog ; 17(3): e1009356, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33647064

RESUMO

Several arenaviruses cause hemorrhagic fevers in humans with high case fatality rates. A vaccine named Candid#1 is available only against Junin virus (JUNV) in Argentina. Specific N-linked glycans on the arenavirus surface glycoprotein (GP) mask important epitopes and help the virus evade antibody responses. However the role of GPC glycans in arenavirus pathogenicity is largely unclear. In a lethal animal model of hemorrhagic fever-causing Machupo virus (MACV) infection, we found that a chimeric MACV with the ectodomain of GPC from Candid#1 vaccine was partially attenuated. Interestingly, mutations resulting in acquisition of N-linked glycans at GPC N83 and N166 frequently occurred in late stages of the infection. These glycosylation sites are conserved in the GPC of wild-type MACV, indicating that this is a phenotypic reversion for the chimeric MACV to gain those glycans crucial for infection in vivo. Further studies indicated that the GPC mutant viruses with additional glycans became more resistant to neutralizing antibodies and more virulent in animals. On the other hand, disruption of these glycosylation sites on wild-type MACV GPC rendered the virus substantially attenuated in vivo and also more susceptible to antibody neutralization, while loss of these glycans did not affect virus growth in cultured cells. We also found that MACV lacking specific GPC glycans elicited higher levels of neutralizing antibodies against wild-type MACV. Our findings revealed the critical role of specific glycans on GPC in arenavirus pathogenicity and have important implications for rational design of vaccines against this group of hemorrhagic fever-causing viruses.


Assuntos
Anticorpos Antivirais/imunologia , Arenavirus/imunologia , Febre Hemorrágica Americana/virologia , Vírus Junin/patogenicidade , Animais , Anticorpos Neutralizantes/imunologia , Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/patogenicidade , Febre Hemorrágica Americana/imunologia , Febre Hemorrágica Americana/prevenção & controle , Humanos , Vírus Junin/imunologia , Vacinas Virais/imunologia
8.
Nat Chem Biol ; 17(10): 1057-1064, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34168368

RESUMO

The predominant approach for antibody generation remains animal immunization, which can yield exceptionally selective and potent antibody clones owing to the powerful evolutionary process of somatic hypermutation. However, animal immunization is inherently slow, not always accessible and poorly compatible with many antigens. Here, we describe 'autonomous hypermutation yeast surface display' (AHEAD), a synthetic recombinant antibody generation technology that imitates somatic hypermutation inside engineered yeast. By encoding antibody fragments on an error-prone orthogonal DNA replication system, surface-displayed antibody repertoires continuously mutate through simple cycles of yeast culturing and enrichment for antigen binding to produce high-affinity clones in as little as two weeks. We applied AHEAD to generate potent nanobodies against the SARS-CoV-2 S glycoprotein, a G-protein-coupled receptor and other targets, offering a template for streamlined antibody generation at large.


Assuntos
Formação de Anticorpos/imunologia , Engenharia de Proteínas/métodos , Proteínas Recombinantes/biossíntese , Anticorpos/imunologia , Antígenos , COVID-19/imunologia , Humanos , Biblioteca de Peptídeos , Proteínas Recombinantes/metabolismo , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Saccharomyces cerevisiae/metabolismo , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/metabolismo , Glicoproteína da Espícula de Coronavírus/imunologia
9.
BMC Bioinformatics ; 23(1): 547, 2022 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-36536276

RESUMO

As of June 2022, the GISAID database contains more than 11 million SARS-CoV-2 genomes, including several thousand nucleotide sequences for the most common variants such as delta or omicron. These SARS-CoV-2 strains have been collected from patients around the world since the beginning of the pandemic. We start by assessing the similarity of all pairs of nucleotide sequences using the Jaccard index and principal component analysis. As shown previously in the literature, an unsupervised cluster analysis applied to the SARS-CoV-2 genomes results in clusters of sequences according to certain characteristics such as their strain or their clade. Importantly, we observe that nucleotide sequences of common variants are often outliers in clusters of sequences stemming from variants identified earlier on during the pandemic. Motivated by this finding, we are interested in applying outlier detection to nucleotide sequences. We demonstrate that nucleotide sequences of common variants (such as alpha, delta, or omicron) can be identified solely based on a statistical outlier criterion. We argue that outlier detection might be a useful surveillance tool to identify emerging variants in real time as the pandemic progresses.


Assuntos
COVID-19 , Humanos , Sequência de Bases , SARS-CoV-2 , Análise por Conglomerados , Bases de Dados Factuais
10.
J Virol ; 95(17): e0186820, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34132574

RESUMO

Pathogenic clade B New World mammarenaviruses (NWM) can cause Argentine, Venezuelan, Brazilian, and Bolivian hemorrhagic fevers. Sequence variability among NWM glycoproteins (GP) poses a challenge to the development of broadly neutralizing therapeutics against the entire clade of viruses. However, blockade of their shared binding site on the apical domain of human transferrin receptor 1 (hTfR1/CD71) presents an opportunity for the development of effective and broadly neutralizing therapeutics. Here, we demonstrate that the murine monoclonal antibody OKT9, which targets the apical domain of hTfR1, can sterically block cellular entry by viral particles presenting clade B NWM glycoproteins (GP1-GP2). OKT9 blockade is also effective against viral particles pseudotyped with glycoproteins of a recently identified pathogenic Sabia-like virus. With nanomolar affinity for hTfR1, the OKT9 antigen binding fragment (OKT9-Fab) sterically blocks clade B NWM-GP1s and reduces infectivity of an attenuated strain of Junin virus. Binding of OKT9 to the hTfR1 ectodomain in its soluble, dimeric state produces stable assemblies that are observable by negative-stain electron microscopy. A model of the OKT9-sTfR1 complex, informed by the known crystallographic structure of sTfR1 and a newly determined structure of the OKT9 antigen binding fragment (Fab), suggests that OKT9 and the Machupo virus GP1 share a binding site on the hTfR1 apical domain. The structural basis for this interaction presents a framework for the design and development of high-affinity, broadly acting agents targeting clade B NWMs. IMPORTANCE Pathogenic clade B NWMs cause grave infectious diseases, the South American hemorrhagic fevers. Their etiological agents are Junin (JUNV), Guanarito (GTOV), Sabiá (SABV), Machupo (MACV), Chapare (CHAV), and a new Sabiá-like (SABV-L) virus recently identified in Brazil. These are priority A pathogens due to their high infectivity and mortality, their potential for person-to-person transmission, and the limited availability of effective therapeutics and vaccines to curb their effects. While low homology between surface glycoproteins of NWMs foils efforts to develop broadly neutralizing therapies targeting NWMs, this work provides structural evidence that OKT9, a monoclonal antibody targeting a single NWM glycoprotein binding site on hTfR1, can efficiently prevent their entry into cells.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Arenavirus do Novo Mundo/fisiologia , Glicoproteínas/imunologia , Febre Hemorrágica Americana/prevenção & controle , Receptores da Transferrina/imunologia , Células A549 , Sequência de Aminoácidos , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Febre Hemorrágica Americana/imunologia , Febre Hemorrágica Americana/virologia , Humanos , Estrutura Terciária de Proteína , Receptores da Transferrina/química , Receptores da Transferrina/genética
14.
J Gen Intern Med ; 34(7): 1184-1191, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30963439

RESUMO

BACKGROUND: Outpatient primary care experience is vital to internal medicine resident training but may impact quality and equity of care delivered in practices that include resident physicians. Understanding whether quality differences exist among resident and staff primary care physicians (PCPs) may present an opportunity to address health disparities within academic medical centers. OBJECTIVE: To determine whether there are differences in the quality of primary care provided by resident PCPs compared to staff PCPs. DESIGN: A retrospective cohort study with a propensity-matched analysis. PARTICIPANTS: 143,274 patients, including 10,870 patients managed by resident PCPs, seen in 16 primary care practices affiliated with an academic medical center. MAIN MEASURES: Guideline-concordant chronic disease management of diabetes (HbA1c, LDL) and coronary artery disease (LDL), preventive breast, cervical, and colorectal cancer screening, and resource utilization measures including emergency department (ED) visits, hospitalizations, high-cost imaging, and patient-reported health experience. KEY RESULTS: At baseline, there were significant differences in sociodemographic and clinical characteristics between resident and staff physician patients. Resident patients were less likely to achieve chronic disease and preventive cancer screening outcome measures including LDL at goal (adjusted OR [aOR] 0.77 [95% CI 0.65, 0.92]) for patients with coronary artery disease; HbA1c at goal (aOR 0.73 [95% CI 0.62, 0.85]) for patients with diabetes; breast (aOR 0.56 [95% CI 0.49, 0.63]), cervical (aOR 0.66 [95% CI 0.60, 0.74]), and colorectal (aOR 0.72 [95% CI 0.65, 0.79] cancer screening. Additionally, resident patients had higher rates of ED visits and hospitalizations but lower rates of high-cost imaging. Resident patients reported lower rates of satisfaction with certain access to care and communication measures. Similar outcomes were noted in propensity-matched sensitivity analyses. CONCLUSION: After controlling for differences in sociodemographic and clinical factors, resident patients were less likely to achieve chronic disease and preventive cancer screening outcomes compared to staff patients. Further efforts to address ambulatory trainee education and primary care quality along with novel approaches to the management of the disproportionately disadvantaged resident patient panels are needed.


Assuntos
Equidade em Saúde/normas , Internato e Residência/normas , Medidas de Resultados Relatados pelo Paciente , Médicos de Atenção Primária/normas , Atenção Primária à Saúde/normas , Qualidade da Assistência à Saúde/normas , Adulto , Estudos de Coortes , Feminino , Humanos , Internato e Residência/métodos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Atenção Primária à Saúde/métodos , Estudos Retrospectivos
16.
PLoS Biol ; 11(5): e1001571, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23723737

RESUMO

Transferrin Receptor (TfR1) is the cell-surface receptor that regulates iron uptake into cells, a process that is fundamental to life. However, TfR1 also facilitates the cellular entry of multiple mammalian viruses. We use evolutionary and functional analyses of TfR1 in the rodent clade, where two families of viruses bind this receptor, to mechanistically dissect how essential housekeeping genes like TFR1 successfully balance the opposing selective pressures exerted by host and virus. We find that while the sequence of rodent TfR1 is generally conserved, a small set of TfR1 residue positions has evolved rapidly over the speciation of rodents. Remarkably, all of these residues correspond to the two virus binding surfaces of TfR1. We show that naturally occurring mutations at these positions block virus entry while simultaneously preserving iron-uptake functionalities, both in rodent and human TfR1. Thus, by constantly replacing the amino acids encoded at just a few residue positions, TFR1 divorces adaptation to ever-changing viruses from preservation of key cellular functions. These dynamics have driven genetic divergence at the TFR1 locus that now enforces species-specific barriers to virus transmission, limiting both the cross-species and zoonotic transmission of these viruses.


Assuntos
Genes Essenciais , Receptores da Transferrina/metabolismo , Sequência de Aminoácidos , Animais , Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/patogenicidade , Linhagem Celular , Cães , Humanos , Ferro/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação , Polimorfismo de Nucleotídeo Único , Receptores da Transferrina/genética , Receptores Virais/química , Receptores Virais/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Internalização do Vírus , Zoonoses
17.
Expert Rev Endocrinol Metab ; 19(4): 385-391, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38567714

RESUMO

BACKGROUND: We investigated how a personalized care-planning software and linked mobile-app may aid people to self-manage their type 2 diabetes (T2D) more effectively. RESEARCH DESIGN AND METHODS: People with T2D and glycated hemoglobin (HbA1c) greater than 58 mmol/mol (7.5%) were randomized to either an intervention group receiving a personalized care plan, or the control group receiving usual care. Quality of life (QoL) was measured for both groups using validated questionnaires and one-on-one interviews with a subset of 12 participants from each group. RESULTS: QoL for the active treatment group increased, by their EQ -5D-5 L score increasing on average by 0.046, whereas it decreased for the control group on average by 0.009. The EQ Visual Analogue Score (VAS) of the intervention group also increased by 8.2%, whereas the control group had a reduction in EQ VAS score of 2.8% (p = 0.008 for difference). CONCLUSION: In this prospective RCT, the findings point to how the provision of personalized care plans can result in an improvement in individuals' self-rated QoL. This may lead to longer term health benefits.


Assuntos
Diabetes Mellitus Tipo 2 , Medidas de Resultados Relatados pelo Paciente , Qualidade de Vida , Humanos , Diabetes Mellitus Tipo 2/terapia , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Hemoglobinas Glicadas/análise , Aplicativos Móveis , Estudos Prospectivos , Adulto , Inquéritos e Questionários , Autogestão/métodos
18.
J Virol ; 86(7): 4024-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22278244

RESUMO

Five New World (NW) arenaviruses cause human hemorrhagic fevers. Four of these arenaviruses are known to enter cells by binding human transferrin receptor 1 (hTfR1). Here we show that the fifth arenavirus, Chapare virus, similarly uses hTfR1. We also identify an anti-hTfR1 antibody, ch128.1, which efficiently inhibits entry mediated by the glycoproteins of all five viruses, as well as replication of infectious Junín virus. Our data indicate that all NW hemorrhagic fever arenaviruses utilize a common hTfR1 apical-domain epitope and suggest that therapeutic agents targeting this epitope, including ch128.1 itself, can be broadly effective in treating South American hemorrhagic fevers.


Assuntos
Anticorpos/imunologia , Antígenos CD/química , Antígenos CD/imunologia , Arenavirus do Novo Mundo/fisiologia , Regulação para Baixo , Febres Hemorrágicas Virais/virologia , Receptores da Transferrina/química , Receptores da Transferrina/imunologia , Internalização do Vírus , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Linhagem Celular , Febres Hemorrágicas Virais/genética , Febres Hemorrágicas Virais/imunologia , Humanos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Receptores da Transferrina/genética , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/imunologia , Alinhamento de Sequência
19.
Nature ; 446(7131): 92-6, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17287727

RESUMO

At least five arenaviruses cause viral haemorrhagic fevers in humans. Lassa virus, an Old World arenavirus, uses the cellular receptor alpha-dystroglycan to infect cells. Machupo, Guanarito, Junin and Sabia viruses are New World haemorrhagic fever viruses that do not use alpha-dystroglycan. Here we show a specific, high-affinity association between transferrin receptor 1 (TfR1) and the entry glycoprotein (GP) of Machupo virus. Expression of human TfR1, but not human transferrin receptor 2, in hamster cell lines markedly enhanced the infection of viruses pseudotyped with the GP of Machupo, Guanarito and Junin viruses, but not with those of Lassa or lymphocytic choriomeningitis viruses. An anti-TfR1 antibody efficiently inhibited the replication of Machupo, Guanarito, Junin and Sabia viruses, but not that of Lassa virus. Iron depletion of culture medium enhanced, and iron supplementation decreased, the efficiency of infection by Junin and Machupo but not Lassa pseudoviruses. These data indicate that TfR1 is a cellular receptor for New World haemorrhagic fever arenaviruses.


Assuntos
Antígenos CD/metabolismo , Arenavirus do Novo Mundo/metabolismo , Receptores da Transferrina/metabolismo , Receptores Virais/metabolismo , Anticorpos/imunologia , Anticorpos/farmacologia , Antígenos CD/genética , Antígenos CD/imunologia , Arenavirus do Novo Mundo/efeitos dos fármacos , Arenavirus do Novo Mundo/fisiologia , Meios de Cultura/química , Glicoproteínas/metabolismo , Humanos , Ferro/análise , Ferro/farmacologia , Receptores da Transferrina/antagonistas & inibidores , Receptores da Transferrina/genética , Receptores da Transferrina/imunologia , Proteínas do Envelope Viral/metabolismo , Replicação Viral/efeitos dos fármacos
20.
Diabetes Ther ; 14(6): 977-988, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37079268

RESUMO

INTRODUCTION: Effective and scalable solutions to support management of Type 2 Diabetes (T2D) at a distance are a priority for health systems worldwide. The use of personalised care planning has been shown to be effective at improving the health outcomes and the experience of care amongst people with T2D and other long-term health conditions. Here we describe a specific example of such an intervention. METHODS: The sample comprised 197 participants with T2D randomised to either the active intervention group with digital health planning (App + usual care), with 115 participants, or the control group (usual care), with 82 participants. We analysed data in relation to changes in body mass index (BMI) and glycated haemoglobin (HbA1c) over a 6-month follow-up period. We also analysed responses to questionnaires sent out and held interviews with participants that were in the active treatment group and therefore had a care plan created and access to an app. RESULTS: The active treatment group had significant reductions in HbA1c (p < 0.01) and BMI (p < 0.037) vs the control group (no significant change). The average percentage change in HbA1c for the treatment group over 6 months was - 7.4% (± SE 1.4%), compared with 1.8% (± SE 2.1%) for the control group. The average percentage change in BMI for the treatment group was - 0.7% (± SE 0.4%) and it was - 0.2% (± SE 0.5%) for the control group. A higher percentage of the active treatment group reduced their HbA1c and BMI than the control group. For HbA1c, 72.4% of the active treatment group reduced their HbA1c, compared to 41.5% of the control group. For BMI, 52.7% of the active treatment group experienced a reduction, compared to 42.9% for the control group. Self-measured quality of life (QoL) improved for patients in the active treatment group, shown by an increase in their pre-trial to post-trial EQ-5D-5L rating by an average of 0.0464 (± SE 0.0625), compared to a decrease of 0.0086 (± SE 0.0530) for the control group. The average EQ VAS score also increased pre- to post-trial for the active treatment group, on average by 8.2%, whereas it decreased by an  average of - 2.8% for the control group. CONCLUSION: These findings point to how the provision of personalised plans of care, support and education linked to a mobile app, can result in HbA1c and BMI reduction for many individuals with T2D. The use of a patient management app as well as a personalised care plan also led to an improvement in patient self-rated QoL and engagement.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA