Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Mov Disord ; 37(7): 1394-1404, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35579496

RESUMO

BACKGROUND: Viral induction of neurological syndromes has been a concern since parkinsonian-like features were observed in patients diagnosed with encephalitis lethargica subsequent to the 1918 influenza pandemic. Given the similarities in the systemic responses after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection with those observed after pandemic influenza, there is a question whether a similar syndrome of postencephalic parkinsonism could follow coronavirus disease 2019 infection. OBJECTIVE: The goal of this study was to determine whether prior infection with SARS-CoV-2 increased sensitivity to a mitochondrial toxin known to induce parkinsonism. METHODS: K18-hACE2 mice were infected with SARS-CoV-2 to induce mild-to-moderate disease. After 38 days of recovery, mice were administered a non-lesion-inducing dose of the parkinsonian toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and euthanized 7 days later. Subsequent neuroinflammation and substantia nigra pars compacta (SNpc) dopaminergic (DA) neuron loss were determined and compared with SARS-CoV-2 or MPTP alone. RESULTS: K18-hACE2 mice infected with SARS-CoV-2 or MPTP showed no SNpc DA neuron loss after MPTP. In mice infected and recovered from SARS-CoV-2 infection, MPTP induced a 23% or 19% greater loss of SNpc DA neurons than SARS-CoV-2 or MPTP, respectively (P < 0.05). Examination of microglial activation showed a significant increase in the number of activated microglia in both the SNpc and striatum of the SARS-CoV-2 + MPTP group compared with SARS-CoV-2 or MPTP alone. CONCLUSIONS: Our observations have important implications for long-term public health, given the number of people who have survived SARS-CoV-2 infection, as well as for future public policy regarding infection mitigation. However, it will be critical to determine whether other agents known to increase risk for PD also have synergistic effects with SARS-CoV-2 and are abrogated by vaccination. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
COVID-19 , Influenza Humana , Transtornos Parkinsonianos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/efeitos adversos , Animais , COVID-19/complicações , Modelos Animais de Doenças , Dopamina , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Transtornos Parkinsonianos/induzido quimicamente , SARS-CoV-2 , Tirosina 3-Mono-Oxigenase/metabolismo
2.
RNA Biol ; 19(1): 1-11, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34904915

RESUMO

The role for circulating miRNAs as biomarkers of the COVID-19 disease remains uncertain. We analysed the circulating miRNA profile in twelve COVID-19 patients with moderate-severe disease. This analysis was conducted by performing next generation sequencing (NGS) followed by real-time polymerase chain reaction (RT-qPCR). Compared with healthy controls, we detected significant changes in the circulating miRNA profile of COVID-19 patients. The miRNAs that were significantly altered in all the COVID-19 patients were miR-150-5p, miR-375, miR-122-5p, miR-494-3p, miR-3197, miR-4690-5p, miR-1915-3p, and miR-3652. Infection assays performed using miRNA mimics in HEK-293 T cells determined miR-150-5p to have a crucial role in SARS-CoV-2 infection and this was based on the following data: (i) miR-150-5p mimic lowered in vitro SARS-CoV-2 infection; (ii) miR-150-5p inhibitor reversed the effects of miR-150-5p mimic on SARS-CoV-2 infection of cells; and (iii) a novel miRNA recognition element (MRE) was identified in the coding strand of SARS-CoV-2 nsp10, the expression of which could be inhibited by miR-150-5p mimic. Our findings identified crucial miRNA footprints in COVID-19 patients with moderate-severe disease. A combination of co-transfection and Western blotting experiments also determined the ability of miR-150-5p to inhibit SARS-CoV-2 infection via directly interacting with MRE in the coding strand of nsp10. Our investigation showed that a sharp decline in the miR-150-5p plasma levels in COVID-19 patients may support enhanced SARS-CoV-2 infection. Furthermore, this study provides insight into one possible mechanism by which COVID-19-induced changes to miR-150-5p levels may promote SARS-CoV-2 infection via modulating nsp10 expression.


Assuntos
COVID-19/metabolismo , Regulação Viral da Expressão Gênica , MicroRNAs/metabolismo , SARS-CoV-2/metabolismo , Proteínas Virais Reguladoras e Acessórias/biossíntese , Animais , COVID-19/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Humanos , MicroRNAs/genética , SARS-CoV-2/genética , Células Vero , Proteínas Virais Reguladoras e Acessórias/genética
3.
Apoptosis ; 26(1-2): 96-110, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33387145

RESUMO

Burkitt's lymphoma (BL) is the fastest growing human tumor. Current treatment consists of a multiagent regimen of cytotoxic drugs with serious side effjects including tumor lysis, cardiotoxicity, hepatic impairment, neuropathy, myelosuppression, increased susceptibility to malignancy, and death. Furthermore, therapeutic interventions in areas of BL prevalence are not as feasible as in high-income countries. Therefore, there exists an urgent need to identify new therapies with a safer profile and improved accessibility. Triclosan (TCS), an antimicrobial used in personal care products and surgical scrubs, has gained considerable interest as an antitumor agent due to its interference with fatty acid synthesis. Here, we investigate the antitumor properties and associated molecular mechanisms of TCS in BL-derived BJAB cells. Dose-dependent cell death was observed following treatment with 10-100 µM TCS for 24 h, which was associated with membrane phospholipid scrambling, compromised permeability, and cell shrinkage. TCS-induced cell death was accompanied by elevated intracellular calcium, perturbed redox balance, chromatin condensation, and DNA fragmentation. TCS upregulated Bad expression and downregulated that of Bcl2. Moreover, caspase and JNK MAPK signaling were required for the full apoptotic activity of TCS. In conclusion, this report identifies TCS as an antitumor agent and provides new insights into the molecular mechanisms governing TCS-induced apoptosis in BL cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linfoma de Burkitt/fisiopatologia , Triclosan/farmacologia , Antineoplásicos/análise , Linfoma de Burkitt/genética , Linfoma de Burkitt/metabolismo , Cálcio/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Triclosan/análise , Células Tumorais Cultivadas
4.
Arch Virol ; 163(10): 2633-2643, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29936609

RESUMO

Oncogenic viruses carry an extensive arsenal of oncogenes for hijacking cellular pathways. Notably, variations in oncogenes among tumor-producing viruses give rise to different mechanisms for cellular transformation. Specifically, Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus able to infect and transform a variety of cell types. The oncogenicity of KSHV disseminates from the virus' ability to induce and encode a wide variety of both cellular and viral oncogenes. Such an array of cellular and viral oncogenes enables KSHV to induce the malignant phenotype of a KSHV-associated cancer. Evolutionarily, KSHV has acquired many oncogenic homologues capable of inducing cell proliferation, cell differentiation, cell survival, and immune evasion. Integration between inducing and encoding oncogenes plays a vital role in KSHV pathogenicity. KSHV is alleged to harbor the highest number of potential oncogenes by which a virus promotes cellular transformation and malignancy. Many KSHV inducing/encoding oncogenes are mainly expressed during the latent phase of KSHV infection, a period required for virus establishment of malignant cellular transformation. Elucidation of the exact mechanism(s) by which oncogenes promote KSHV pathogenicity would not only give rise to potential novel therapeutic targets/drugs but would also add to our understanding of cancer biology. The scope of this review is to examine the roles of the most important cellular and viral oncogenes involved in KSHV pathogenicity.


Assuntos
Carcinogênese/genética , Transformação Celular Neoplásica/genética , Genes Virais/genética , Herpesvirus Humano 8/genética , Oncogenes/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/patologia , Diferenciação Celular/genética , Proliferação de Células/genética , Sobrevivência Celular/genética , Transformação Celular Neoplásica/patologia , Humanos , Sarcoma de Kaposi/virologia , Evasão Tumoral/genética
5.
Intervirology ; 60(6): 217-226, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29597230

RESUMO

OBJECTIVES: Kaposi sarcoma-associated herpesvirus (KSHV) glycoprotein B (gB) is expressed on the viral envelope as well as on the cytoplasmic membrane of infected cells. In the current study, we aimed to decipher the impact of membrane-associated gB on adhesion and migration of cells via modulating the expression of cytokines. METHODS: A combination of polymerase chain reaction array, cell adhesion assay, and wound-healing migration assay was conducted to study the influence of the gB-induced cytokines on cell adhesion and migration. RESULTS: Membrane-associated gB was demonstrated to significantly upregulate the expression of IL-1ß and TNF-α. Elevated levels of these cytokines were observed in conditioned medium (CM) collected from gB-expressing cells (gB-CM) compared to CM collected from untransfected cells or cells transfected with empty vector. KSHV gB-induced IL-1ß and TNF-α play a role in the ability of gB-CM to mediate cell adhesion while inhibiting migration. CONCLUSION: Our results provide novel evidence that demonstrates full-length gB expressed on cell membrane to mediate adhesion and inhibit migration of cells not only by autocrine mechanism mediated by RGD-based interactions [Hussein et al.: BMC Cancer 2016; 16: 148], but also by paracrine mechanism mediated by gB-induced IL-1ß and TNF-α.

6.
Arch Virol ; 162(11): 3293-3303, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28707270

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) causes a variety of cancers, including Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman disease (MCD). Host cellular microRNAs (miRNAs) play important post-transcriptional regulatory roles in gene expression and can greatly influence virus-host cell interactions. This study investigated cellular miRNA expression profiles operating in response to early stages of KSHV infection of human Burkitt lymphoma B cells (BJAB). We employed deep sequencing to analyze miRNA expression in KSHV-infected BJAB cells 15 min post infection (PI) and compared this to uninfected BJAB cells. A total of 32 known miRNAs and 28 novel miRNA candidates were differentially expressed in KSHV-infected compared to uninfected BJAB cells. Interestingly, miRNA expression profiles during the early stages of viral infection yielded comparable results when UV-inactivated KSHV was used. The deep sequencing results were further confirmed by performing real-time reverse transcription PCR. The target genes predicted to be regulated by both the known and novel miRNAs are mainly involved in assisting virus entry, inducing critical cell signaling, initiating transcription of immediate early genes, promoting latent infection, and modulating the host immune response. For the first time, we provide insight into the host cellular miRNA expression profiles in response to early stages of KSHV infection of human B cells. Furthermore, this study offers a valuable basis for further investigation on the roles of cellular miRNAs in the KSHV entry process.


Assuntos
Herpesvirus Humano 8/fisiologia , MicroRNAs/metabolismo , Linhagem Celular Tumoral , Regulação da Expressão Gênica/imunologia , Herpesvirus Humano 8/efeitos da radiação , Humanos , MicroRNAs/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcriptoma , Raios Ultravioleta
7.
BMC Cancer ; 16: 148, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26912031

RESUMO

BACKGROUND: Kaposi's sarcoma-associated herpesvirus (KSHV) glycoprotein B (gB) is not only expressed on the envelope of mature virions but also on the surfaces of cells undergoing lytic replication. Among herpesviruses, KSHV gB is the only glycoprotein known to possess the RGD (Arg-Gly-Asp) binding integrin domain critical to mediating cell attachment. Recent studies described gB to also possess a disintegrin-like domain (DLD) said to interact with non-RGD binding integrins. We wanted to decipher the roles of two individually distinct integrin binding domains (RGD versus DLD) within KSHV gB in regulating attachment of cells over cell migration. METHODS: We established HeLa cells expressing recombinant full length gB, gB lacking a functional RGD (gBΔR), and gB lacking a functionally intact DLD (gBΔD) on their cell surfaces. These cells were tested in wound healing assay, Transwell migration assay, and adhesion assay to monitor the ability of the RGD and DLD integrin recognition motifs in gB to mediate migration and attachment of cells. We also used soluble forms of the respective gB recombinant proteins to analyze and confirm their effect on migration and attachment of cells. The results from the above studies were authenticated by the use of imaging, and standard biochemical approaches as Western blotting and RNA silencing using small interfering RNA. RESULTS: The present report provides the following novel findings: (i) gB does not induce cell migration; (ii) RGD domain in KSHV gB is the switch that inhibits the ability of DLD to induce cellular migration thus promoting attachment of cells. CONCLUSIONS: Independently, RGD interactions mediate attachment of cells while DLD interactions regulate migration of cells. However, when both RGD and DLD are functionally present in the same protein, gB, the RGD interaction-induced attachment of cells overshadows the ability of DLD mediated signaling to induce migration of cells. Furthering our understanding of the molecular mechanism of integrin engagement with RGD and DLD motifs within gB could identify promising new therapeutic avenues and research areas to explore.


Assuntos
Glicoproteínas/química , Glicoproteínas/metabolismo , Herpesvirus Humano 8/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Sítios de Ligação , Adesão Celular , Movimento Celular , Proliferação de Células , Glicoproteínas/genética , Células HeLa , Herpesvirus Humano 8/química , Herpesvirus Humano 8/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Estrutura Terciária de Proteína , Transdução de Sinais , Proteínas do Envelope Viral/genética
8.
South Med J ; 109(11): 712-717, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27812717

RESUMO

OBJECTIVES: Treatment of human immunodeficiency virus (HIV)-infected patients with tenofovir disoproxil fumarate is associated with a decrease in bone mineral density (BMD). Treatment with efavirenz is associated with vitamin D deficiency. We compared the effects of efavirenz, emtricitabine, and tenofovir disoproxil fumarate (EFV/FTC/TDF) with the effects of raltegravir, darunavir, and ritonavir (RAL/DRV/r) on BMD and 25-hydroxyvitamin D (25[OH]D) levels in HIV-infected, antiretroviral treatment-naïve African American subjects. METHODS: This was a pilot study at a single HIV clinic. Forty HIV treatment-naïve African American subjects were screened, 35 of whom were randomized to receive either EFV/FTC/TDF or RAL/DRV/r. All of the subjects received supplemental vitamin D3 and calcium. CD4 counts, HIV RNA, parathyroid hormone, osteocalcin, N-telopeptide, and 25(OH)D levels were obtained at baseline and at 8, 24, 36, and 48 weeks. Dual-energy x-ray absorptiometry of the spine and hip was performed at baseline and at week 48. RESULTS: Of the 35 subjects enrolled, 10 patients receiving each regimen completed the study. Median baseline 25(OH)D levels were decreased and similar in both groups. All of the patients had plasma HIV RNA <50 copies per milliliter by week 24. By week 48, there was a sustained increase in 25(OH)D in the RAL/DRV/r group (P = 0.0004) but not in the EFV/FTC/TDF group (P = 0.78). There were reductions in BMD of the mean total hip (P = 0.002) and the mean femoral neck (P = 0.004) in the EFV/FTC/TDF group but not in the RAL/DRV/r group. CONCLUSIONS: Treatment of African American patients with HIV using EFV/FTC/TDF is associated with a reduction in BMD of the hip and sustained reductions of 25(OH)D not seen in the group that received RAL/DRV/r. This phenomenon may have long-term consequences on bone integrity in this population.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Densidade Óssea/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , Vitamina D/análogos & derivados , Absorciometria de Fóton , Adulto , Negro ou Afro-Americano , Alcinos , Benzoxazinas/uso terapêutico , Conservadores da Densidade Óssea/administração & dosagem , Contagem de Linfócito CD4 , Cálcio da Dieta/administração & dosagem , Colágeno Tipo I/sangue , Ciclopropanos , Darunavir/uso terapêutico , Quimioterapia Combinada , Emtricitabina/uso terapêutico , Feminino , Colo do Fêmur/diagnóstico por imagem , Articulação do Quadril/diagnóstico por imagem , Humanos , Masculino , Osteocalcina/sangue , Hormônio Paratireóideo/sangue , Peptídeos/sangue , Projetos Piloto , RNA Viral/sangue , Raltegravir Potássico/uso terapêutico , Ritonavir/uso terapêutico , Tenofovir/uso terapêutico , Vitamina D/administração & dosagem , Vitamina D/sangue , Adulto Jovem
9.
Arch Virol ; 160(11): 2669-81, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26321473

RESUMO

Viruses successfully infect host cells by initially binding to the surfaces of the cells, followed by an intricate entry process. As multifunctional heterodimeric cell-surface receptor molecules, integrins have been shown to usefully serve as entry receptors for a plethora of viruses. However, the exact role(s) of integrins in viral pathogen internalization has yet to be elaborately described. Notably, several viruses harbor integrin-recognition motifs displayed on viral envelope/capsid-associated proteins. The most common of these motifs is the minimal peptide sequence for binding integrins, RGD (Arg-Gly-Asp), which is known for its role in virus infection via its ability to interact with over half of the more than 20 known integrins. Not all virus-integrin interactions are RGD-dependent, however. Non-RGD-binding integrins have also been shown to effectively promote virus entry and infection as well. Such virus-integrin binding is shown to facilitate adhesion, cytoskeleton rearrangement, integrin activation, and increased intracellular signaling. Also, we have attempted to discuss the role of carbohydrate moieties in virus interactions with receptor-like host cell surface integrins that drive the process of internalization. As much as possible, this article examines the published literature regarding the role of integrins in terms of virus infection and virus-encoded glycosylated proteins that mediate interactions with integrins, and it explores the idea of targeting these receptors as a therapeutic treatment option.


Assuntos
Integrinas/química , Integrinas/metabolismo , Proteínas do Envelope Viral/metabolismo , Viroses/metabolismo , Viroses/virologia , Vírus/metabolismo , Motivos de Aminoácidos , Animais , Humanos , Integrinas/genética , Ligação Proteica , Proteínas do Envelope Viral/genética , Viroses/genética , Vírus/química , Vírus/genética
10.
J Gen Virol ; 95(Pt 8): 1770-1782, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24814923

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) glycoprotein B (gB) is a lytic structural protein expressed on the envelope of mature virions and on the membrane of cells supporting lytic infection. In addition to this viral glycoprotein's interaction with integrins via its RGD (Arg-Gly-Asp) motif, KSHV gB possesses a disintegrin-like domain (DLD), which binds integrins as well. Prior to this study, there has been minimal research involving the less common integrin-binding motif, DLD, of gB as it pertains to herpesvirus infection. By using phage display peptide library screening and molecular biology techniques, the DLD of KSHV gB was shown to interact specifically with non-RGD binding α9ß1 integrins. Similarly, monitoring wild-type infection confirmed α9ß1:DLD interactions to be critical to successful KSHV infection of human foreskin fibroblast (HFF) cells and human dermal microvascular endothelial cells (HMVEC-d) compared with 293 cells. To further demonstrate the importance of the DLD of gB in KSHV infection, two recombinant virus constructs were generated using a bacterial artificial chromosome (BAC) system harbouring the KSHV genome (BAC36): BAC36ΔD-KSHV (lacking a functionally intact DLD of gB and containing an introduced tetracycline cassette) and BAC36.T-KSHV (containing an intact DLD sequence and an introduced tetracycline cassette). Accordingly, BAC36ΔD-KSHV presented significantly lower infection rates in HFF and HMVEC-d cells compared with the comparable infection rates achieved by wild-type BAC36-KSHV and BAC36.T-KSHV. Thus, the present report has delineated a critical role for the DLD of gB in KSHV infection, which may lead to a broader knowledge regarding the sophisticated mechanisms utilized by virus-encoded structural proteins in KSHV entry and infection.


Assuntos
Herpesvirus Humano 8/fisiologia , Interações Hospedeiro-Patógeno , Integrinas/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Células Cultivadas , Células Endoteliais/virologia , Fibroblastos/virologia , Humanos , Estrutura Terciária de Proteína
11.
Toxics ; 12(7)2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-39058147

RESUMO

Smart Nano-enabled Antiviral Therapeutic (SNAT) is a promising nanodrug that previously demonstrated efficacy in preclinical studies to alleviate SARS-CoV-2 pathology in hamsters. SNAT comprises taxoid (Tx)-decorated amino (NH2)-functionalized near-atomic size positively charged silver nanoparticles (Tx-[NH2-AgNPs]). Herein, we aimed to elucidate the molecular mechanism of the viral inhibition and safety of aerosolized SNAT treatment in SARS-CoV-2-infected golden Syrian hamsters. High-resolution transmission electron microscopy (HR-TEM) coupled with energy dispersive spectroscopy (EDS) and ELISAs showed SNAT binds directly to the SARS-CoV-2 virus by interacting with intact spike (S) protein, specifically to S2 subunit. SNAT (≥1 µg/mL) treatment significantly lowered SARS-CoV-2 infections of Calu-3 cells. Extraction-free whole transcriptome assay was used to detect changes in circulatory micronome in hamsters treated intranasally with SNAT (two doses of 10 µg/mL of 2 mL each administered 24 h apart). Uninfected hamsters treated with SNAT had altered circulatory concentrations of 18 microRNAs (8 miRNAs upregulated, 10 downregulated) on day 3 post-treatment compared to uninfected controls. SNAT-induced downregulation of miR-141-3p and miR-200b-3p may reduce viral replication and inflammation by targeting Ythdf2 and Slit2, respectively. Further, SNAT treatment significantly lowered IL-6 expression in infected hamster lungs compared to untreated infected hamsters. Taken together, we demonstrate that SNAT binds directly to SARS-CoV-2 via the S protein to prevent viral entry and propose a model by which SNAT alters the cellular miRNA-directed milieu to promote antiviral cellular processes and neutralize infection. Our results provide insights into the use of low-dose intranasally delivered SNAT in treating SARS-CoV-2 infections in a hamster model.

12.
Sci Total Environ ; 925: 171675, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38485022

RESUMO

Globally rising antibiotic-resistant (AR) and multi-drug resistant (MDR) bacterial infections are of public health concern due to treatment failure with current antibiotics. Enterobacteria, particularly Escherichia coli, cause infections of surgical wound, bloodstream, and urinary tract, including pneumonia and sepsis. Herein, we tested in vitro antibacterial efficacy, mode of action (MoA), and safety of novel amino-functionalized silver nanoparticles (NH2-AgNP) against the AR bacteria. Two AR E. coli strains (i.e., ampicillin- and kanamycin-resistant E. coli), including a susceptible strain of E. coli DH5α, were tested for susceptibility to NH2-AgNP using Kirby-Bauer disk diffusion and standard growth assays. Dynamic light scattering (DLS) was used to determine cell debris and relative conductance was used as a measure of cell leakage, and results were confirmed with transmission electron microscopy (TEM). Multiple oxidative stress assays were used for in vitro safety evaluation of NH2-AgNP in human lung epithelial cells. Results showed that ampicillin and kanamycin did not inhibit growth in either AR bacterial strain with doses up to 160 µg/mL tested. NH2-AgNP exhibited broad-spectrum bactericidal activity, inhibiting the growth of all three bacterial strains at doses ≥1 µg/mL. DLS and TEM revealed cell debris formation and cell leakage upon NH2-AgNP treatment, suggesting two possible MoAs: electrostatic interactions followed by cell wall damage. Safety evaluation revealed NH2-AgNP as noncytotoxic and antioxidative to human lung epithelial cells. Taken together, these results suggest that NH2-AgNP may serve as an effective and safer bactericidal therapy against AR bacterial infections compared to common antibiotics.


Assuntos
Infecções Bacterianas , Nanopartículas Metálicas , Humanos , Antibacterianos/toxicidade , Escherichia coli , Prata/toxicidade , Nanopartículas Metálicas/toxicidade , Bactérias , Ampicilina/farmacologia , Canamicina/farmacologia , Testes de Sensibilidade Microbiana
13.
Sci Total Environ ; 906: 167492, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37778563

RESUMO

Arguably the most ecologically and economically valuable pollinators worldwide, honey bees play a significant role in food production and enrich biodiversity through pollination. Varroa destructor is an invasive ectoparasitic mite that attacks and feeds on European honey bee, Apis mellifera. Because literature on the effectiveness and sustainability of various treatment modalities available for Varroa mite control in honey bee colonies are scattered, this scoping review was conducted to serve as a guiding document with a focus on: (1) identifying the detrimental impact Varroa mites have on the European honey bee; (2) determining current methods for Varroa mite control and their limitations; (3) examining current market landscape and key players in the pesticide market; and (4) identifying opportunities for more sustainable Varroa mite control methods. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines, 397 articles published between 1998 and 2022 were screened; of which 65 articles were retained using inclusion/exclusion criteria, which were systematically analyzed in-depth, information extracted, and included in this scoping review. The results suggest that Varroa mites are one of the predominant causes of global honey bee decline as they lack natural resistance to Varroa mites, thereby negatively affecting honey bee reproduction and immunity, killing broods, and transmitting pathogenic viruses to colonies. Further, our findings suggest that: apiarists have many options for Varroa control, but no method has proven to be effective, safe and nonpersistent in the environment; adoption of nano-pesticides and development of sustainable alternatives to traditional pesticides are key drivers for growing pesticide market; and nano-pesticides may have potential to serve as an effective, safe and non-ecopersistent pesticide for Varroa mite and associated virus control. In conclusion, this review highlights an unmet need for effective and sustainable control strategies and tools for Varroa mite and virus control.


Assuntos
Praguicidas , Varroidae , Abelhas , Animais , Imunidade Inata , Interações Hospedeiro-Parasita
14.
Biology (Basel) ; 12(10)2023 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-37887044

RESUMO

Since the discovery of microRNAs (miRNAs) in C. elegans in 1993, the field of miRNA research has grown steeply. These single-stranded non-coding RNA molecules canonically work at the post-transcriptional phase to regulate protein expression. miRNAs are known to regulate viral infection and the ensuing host immune response. Evolving research suggests miRNAs are assets in the discovery and investigation of therapeutics and diagnostics. In this review, we succinctly summarize the latest findings in (i) mechanisms underpinning miRNA regulation of viral infection, (ii) miRNA regulation of host immune response to viral pathogens, (iii) miRNA-based diagnostics and therapeutics targeting viral pathogens and challenges, and (iv) miRNA patents and the market landscape. Our findings show the differential expression of miRNA may serve as a prognostic biomarker for viral infections in regard to predicting the severity or adverse health effects associated with viral diseases. While there is huge market potential for miRNA technology, the novel approach of using miRNA mimics to enhance antiviral activity or antagonists to inhibit pro-viral miRNAs has been an ongoing research endeavor. Significant hurdles remain in terms of miRNA delivery, stability, efficacy, safety/tolerability, and specificity. Addressing these challenges may pave a path for harnessing the full potential of miRNAs in modern medicine.

15.
Cells ; 12(16)2023 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-37626917

RESUMO

Evidence suggests that patients with long COVID can experience neuropsychiatric, neurologic, and cognitive symptoms. However, these clinical data are mostly associational studies complicated by confounding variables, thus the mechanisms responsible for persistent symptoms are unknown. Here we establish an animal model of long-lasting effects on the brain by eliciting mild disease in K18-hACE2 mice. Male and female K18-hACE2 mice were infected with 4 × 103 TCID50 of SARS-CoV-2 and, following recovery from acute infection, were tested in the open field, zero maze, and Y maze, starting 30 days post infection. Following recovery from SARS-CoV-2 infection, K18-hACE2 mice showed the characteristic lung fibrosis associated with SARS-CoV-2 infection, which correlates with increased expression of the pro-inflammatory kinin B1 receptor (B1R). These mice also had elevated expression of B1R and inflammatory markers in the brain and exhibited behavioral alterations such as elevated anxiety and attenuated exploratory behavior. Our data demonstrate that K18-hACE2 mice exhibit persistent effects of SARS-CoV-2 infection on brain tissue, revealing the potential for using this model of high sensitivity to SARS-CoV-2 to investigate mechanisms contributing to long COVID symptoms in at-risk populations. These results further suggest that elevated B1R expression may drive the long-lasting inflammatory response associated with SARS-CoV-2 infection.


Assuntos
COVID-19 , Feminino , Masculino , Animais , Humanos , Camundongos , COVID-19/complicações , Síndrome de COVID-19 Pós-Aguda , SARS-CoV-2 , Doenças Neuroinflamatórias , Cininas
16.
Sci Rep ; 12(1): 6729, 2022 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-35468937

RESUMO

To tackle growing antibiotic resistance (AR) and hospital-acquired infections (HAIs), novel antimicrobials are warranted that are effective against HAIs and safer for human use. We hypothesize that small 5 nm size positively charged nanoparticles could specifically target bacterial cell wall and adherent fimbriae expression, serving as the next generation antibacterial agent. Herein we show highly positively charged, 5 nm amino-functionalized silver nanoparticles (NH2-AgNPs) were bactericidal; highly negatively charged, 45 nm citrate-functionalized AgNPs (Citrate-AgNPs) were nontoxic; and Ag+ ions were bacteriostatic forming honeycomb-like potentially resistant phenotype, at 10 µg Ag/mL in E. coli. Further, adherent fimbriae were expressed with Citrate-AgNPs (0.5-10 µg/mL), whereas NH2-AgNPs (0.5-10 µg/mL) or Ag+ ions (only at 10 µg/mL) inhibited fimbriae expression. Our results also showed no lipid peroxidation in human lung epithelial and dermal fibroblast cells upon NH2-AgNPs treatments, suggesting NH2-AgNPs as a biocompatible antibacterial candidate. Potent bactericidal effects demonstrated by biocompatible NH2-AgNPs and the lack of toxicity of Citrate-AgNPs lend credence to the hypothesis that small size, positively charged AgNPs may serve as a next-generation antibacterial agent, potentially addressing the rising HAIs and patient health and safety.


Assuntos
Nanopartículas Metálicas , Prata , Antibacterianos/farmacologia , Parede Celular , Ácido Cítrico/farmacologia , Di-Hidrotaquisterol/farmacologia , Escherichia coli , Humanos , Íons/farmacologia , Prata/farmacologia
17.
Drug Deliv Transl Res ; 12(12): 3007-3016, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35441321

RESUMO

To address the unprecedented global public health crisis due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we designed and developed a novel antiviral nano-drug, called SNAT (Smart Nano-Enabled Antiviral Therapeutic), comprised of taxoid (Tx)-decorated amino (NH2)-functionalized near-atomic size positively charged silver nanoparticles (Tx-[NH2-AgNPs]) that are stable for over 3 years. Using a hamster model, we tested the preclinical efficacy of inhaled SNAT on the body weight, virus titer, and histopathology of lungs in SARS-CoV-2-infected hamsters, including biocompatibility in human lung epithelium and dermal fibroblasts using lactase dehydrogenase (LDH) and malondialdehyde (MDA) assays. Our results showed SNAT could effectively reverse the body weight loss, reduce the virus load in oral swabs, and improve lung health in hamsters. Furthermore, LDH assay showed SNAT is noncytotoxic, and MDA assay demonstrated SNAT to be an antioxidant, potentially quenching lipid peroxidation, in both the human cells. Overall, these promising pilot preclinical findings suggest SNAT as a novel, safer antiviral drug lead against SARS-CoV-2 infection and may find applications as a platform technology against other respiratory viruses of epidemic and pandemic potential.


Assuntos
Tratamento Farmacológico da COVID-19 , Nanopartículas Metálicas , Cricetinae , Animais , Humanos , SARS-CoV-2 , Modelos Animais de Doenças , Prata , Antivirais/farmacologia , Antivirais/uso terapêutico
18.
Viruses ; 14(12)2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36560686

RESUMO

Wide variability exists with host response to SARS-CoV-2 infection among individuals. Circulatory micro RNAs (miRNAs) are being recognized as promising biomarkers for complex traits, including viral pathogenesis. We hypothesized that circulatory miRNAs at 48 h post hospitalization may predict the length of stay (LOS) and prognosis of COVID-19 patients. Plasma miRNA levels were compared between three groups: (i) healthy volunteers (C); (ii) COVID-19 patients treated with remdesivir (an antiviral) plus dexamethasone (a glucocorticoid) (with or without baricitinib, a Janus kinase inhibitor) on the day of hospitalization (I); and COVID-19 patients at 48 h post treatment (T). Results showed that circulatory miR-6741-5p expression levels were significantly different between groups C and I (p < 0.0000001); I and T (p < 0.0000001); and C and T (p = 0.001). Our ANOVA model estimated that all patients with less than 12.42 Log2 CPM had a short LOS, or a good prognosis, whereas all patients with over 12.42 Log2 CPM had a long LOS, or a poor prognosis. In sum, we show that circulatory miR-6741-5p may serve as a prognostic biomarker effectively predicting mortality risk and LOS of hospitalized COVID-19 patients.


Assuntos
COVID-19 , MicroRNAs , Humanos , Tempo de Internação , Prognóstico , COVID-19/diagnóstico , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , MicroRNAs/metabolismo , Biomarcadores
19.
Cells ; 11(5)2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35269416

RESUMO

The TP53 tumor suppressor is mutated in ~75% of pancreatic cancers. The mutant TP53 protein in pancreatic ductal adenocarcinomas (PDAC) promotes tumor growth and metastasis. Attempts have been made to develop molecules that restore at least some of the properties of wild-type (WT) TP53. APR-246 is one such molecule, and it is referred to as a mutant TP53 reactivator. To understand the potential of APR-246 to sensitize PDAC cells to chemotherapy, we introduced a vector encoding WT-TP53 into two PDAC cell lines, one lacking the expression of TP53 (PANC-28) and one with a gain-of-function (GOF) mutant TP53 (MIA-PaCa-2). APR-246 increased drug sensitivity in the cells containing either a WT or mutant TP53 protein with GOF activity, but not in cells that lacked TP53. The introduction of WT-T53 into PANC-28 cells increased their sensitivity to the TP53 reactivator, chemotherapeutic drugs, and signal transduction inhibitors. The addition of WT-TP53 to PDAC cells with GOF TP53 also increased their sensitivity to the drugs and therapeutics, indicating that APR-246 could function in cells with WT-TP53 and GOF TP53. These results highlight the importance of knowledge of the type of TP53 mutation that is present in cancer patients before the administration of drugs which function through the reactivation of TP53.


Assuntos
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Adenocarcinoma/tratamento farmacológico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Linhagem Celular Tumoral , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Quinuclidinas/uso terapêutico , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias Pancreáticas
20.
Adv Biol Regul ; 83: 100840, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34866036

RESUMO

Approaches to improve pancreatic cancer therapy are essential as this disease has a very bleak outcome. Approximately 80% of pancreatic cancers are pancreatic ductal adenocarcinomas (PDAC). A key regulatory gene frequently mutated (∼75%) in PDAC is the TP53 tumor suppressor gene which controls the transcription of multiple genes involved in cell cycle progression, apoptosis, cancer progression and other growth regulatory processes. The mouse double minute 2 homolog (MDM2) gene product is a nuclear-localized E3 ubiquitin ligase and negatively regulates the TP53 protein which results in its proteasomal degradation. Various MDM2 inhibitors have been isolated and examined in clinical trials, especially in patients with hematological malignancies. Nutlin-3a is one of the first MDM2 inhibitors isolated. Berberine (BBR) is a natural product found in many fruits and berries and used in traditional medicine for centuries. It has many biological effects, and some are anti-proliferative in nature. BBR may activate the expression of TP53 and inhibit cell cycle progression as well as other events important in cell growth. To understand more about the potential of compounds like BBR and chemical modified BBRs (NAX compounds) to sensitize PDAC cells to MDM2 inhibitors, we introduced either WT-TP53 or the pLXSN empty vector control into two PDAC cell lines, one lacking expression of TP53 (PANC-28) and one with gain-of-function mutant TP53 on both alleles (MIA-PaCa-2). Our results indicate that nutlin-3a was able to increase the sensitivity to BBR and certain NAX compounds. The effects of nutlin-3a were usually more substantial in those cells containing an introduced WT TP53 gene. These results highlight the importance of knowledge of the type of TP53 mutation that is present in cancer patients before the administration of drugs which function by stabilization of the TP53 protein.


Assuntos
Berberina , Neoplasias Pancreáticas , Apoptose , Berberina/farmacologia , Berberina/uso terapêutico , Linhagem Celular Tumoral , Humanos , Imidazóis , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Piperazinas , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA