Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Sci Rep ; 12(1): 857, 2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35039591

RESUMEN

Sorghum damping-off, caused by Fusarium solani (Mart.) Sacc., is a serious disease which causes economic loss in sorghum production. In this study, antagonistic activity of lavender essential oil (EO) at 0.5, 0.75, 1.0, 1.25, 1.5, and 1.6% against F. solani was studied in vitro. Their effects on regulation of three SbWRKY transcription factors, the response factor JERF3 and eight defense-related genes, which mediate different signaling pathways, in sorghum were investigated. Effects of application under greenhouse conditions were also evaluated. The results showed that lavender EO possesses potent antifungal activity against F. solani. A complete inhibition in the fungal growth was recorded for lavender EO at 1.6%. Gas chromatography-mass spectrometric analysis revealed that EO antifungal activity is most likely attributed to linalyl anthranilate, α-terpineol, eucalyptol, α-Pinene, and limonene. Observations using transmission electron microscopy revealed many abnormalities in the ultrastructures of the fungal mycelium as a response to treating with lavender EO, indicating that multi-mechanisms contributed to their antagonistic behavior. Results obtained from Real-time PCR investigations demonstrated that the genes studied were overexpressed, to varying extents in response to lavender EO. However, SbWRKY1 was the highest differentially expressed gene followed by JERF3, which suggest they play primary role(s) in synchronously organizing the transcription-regulatory-networks enhancing the plant resistance. Under greenhouse conditions, treating of sorghum grains with lavender EO at 1.5% prior to infection significantly reduced disease severity. Moreover, the growth parameters evaluated, the activities of antioxidant enzymes, and total phenolic and flavonoid contents were all enhanced. In contrast, lipid peroxidation was highly reduced. Results obtained from this study support the possibility of using lavender EO for control of sorghum damping-off. However, field evaluation is highly needed prior to any usage recommendation.


Asunto(s)
Antifúngicos , Fusarium/efectos de los fármacos , Fusarium/patogenicidad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Expresión Génica/efectos de los fármacos , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Lavandula/química , Aceites Volátiles/farmacología , Enfermedades de las Plantas/genética , Enfermedades de las Plantas/microbiología , Aceites de Plantas/farmacología , Sorghum/genética , Sorghum/microbiología , Factores de Transcripción/genética , Farmacorresistencia Fúngica , Expresión Génica/genética , Redes Reguladoras de Genes/efectos de los fármacos , Redes Reguladoras de Genes/genética , Aceites Volátiles/aislamiento & purificación , Aceites de Plantas/aislamiento & purificación , Factores de Transcripción/metabolismo
2.
Viruses ; 13(10)2021 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-34696360

RESUMEN

Pepper mottle virus (PepMoV) is a destructive pathogen that infects various solanaceous plants, including pepper, bell pepper, potato, and tomato. In this review, we summarize what is known about the molecular characteristics of PepMoV and its interactions with host plants. Comparisons of symptom variations caused by PepMoV isolates in plant hosts indicates a possible relationship between symptom development and genetic variation. Researchers have investigated the PepMoV-plant pathosystem to identify effective and durable genes that confer resistance to the pathogen. As a result, several recessive pvr or dominant Pvr resistance genes that confer resistance to PepMoV in pepper have been characterized. On the other hand, the molecular mechanisms underlying the interaction between these resistance genes and PepMoV-encoded genes remain largely unknown. Our understanding of the molecular interactions between PepMoV and host plants should be increased by reverse genetic approaches and comprehensive transcriptomic analyses of both the virus and the host genes.


Asunto(s)
Interacciones Microbiota-Huesped , Enfermedades de las Plantas/virología , Potyvirus/fisiología , Genes prv , Interacciones Microbiota-Huesped/genética , Solanum lycopersicum/genética , Solanum lycopersicum/virología , Enfermedades de las Plantas/genética , Potyvirus/genética , Solanum tuberosum/genética , Solanum tuberosum/virología
3.
PLoS Comput Biol ; 17(9): e1009418, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34555024

RESUMEN

Increasing body of experimental evidence suggests that anticancer and antimicrobial therapies may themselves promote the acquisition of drug resistance by increasing mutability. The successful control of evolving populations requires that such biological costs of control are identified, quantified and included to the evolutionarily informed treatment protocol. Here we identify, characterise and exploit a trade-off between decreasing the target population size and generating a surplus of treatment-induced rescue mutations. We show that the probability of cure is maximized at an intermediate dosage, below the drug concentration yielding maximal population decay, suggesting that treatment outcomes may in some cases be substantially improved by less aggressive treatment strategies. We also provide a general analytical relationship that implicitly links growth rate, pharmacodynamics and dose-dependent mutation rate to an optimal control law. Our results highlight the important, but often neglected, role of fundamental eco-evolutionary costs of control. These costs can often lead to situations, where decreasing the cumulative drug dosage may be preferable even when the objective of the treatment is elimination, and not containment. Taken together, our results thus add to the ongoing criticism of the standard practice of administering aggressive, high-dose therapies and motivate further experimental and clinical investigation of the mutagenicity and other hidden collateral costs of therapies.


Asunto(s)
Farmacorresistencia Microbiana/genética , Resistencia a Antineoplásicos/genética , Antiinfecciosos/administración & dosificación , Antineoplásicos/administración & dosificación , Biología Computacional , Simulación por Computador , Relación Dosis-Respuesta a Droga , Evolución Molecular , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Humanos , Modelos Biológicos , Mutación/efectos de los fármacos , Tasa de Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Fenotipo , Procesos Estocásticos
4.
Nat Commun ; 12(1): 4181, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34234136

RESUMEN

Nucleobase and nucleoside analogs (NNA) are widely used as anti-viral and anti-cancer agents, and NNA phosphorylation is essential for the activity of this class of drugs. Recently, diphosphatase NUDT15 was linked to thiopurine metabolism with NUDT15 polymorphism associated with drug toxicity in patients. Profiling NNA drugs, we identify acyclovir (ACV) and ganciclovir (GCV) as two new NNAs metabolized by NUDT15. NUDT15 hydrolyzes ACV and GCV triphosphate metabolites, reducing their effects against cytomegalovirus (CMV) in vitro. Loss of NUDT15 potentiates cytotoxicity of ACV and GCV in host cells. In hematopoietic stem cell transplant patients, the risk of CMV viremia following ACV prophylaxis is associated with NUDT15 genotype (P = 0.015). Donor NUDT15 deficiency is linked to graft failure in patients receiving CMV-seropositive stem cells (P = 0.047). In conclusion, NUDT15 is an important metabolizing enzyme for ACV and GCV, and NUDT15 variation contributes to inter-patient variability in their therapeutic effects.


Asunto(s)
Aciclovir/farmacología , Antivirales/farmacología , Infecciones por Citomegalovirus/prevención & control , Ganciclovir/análogos & derivados , Pirofosfatasas/genética , Aciclovir/uso terapéutico , Adolescente , Adulto , Anciano , Animales , Profilaxis Antibiótica , Antivirales/uso terapéutico , Variación Biológica Poblacional/genética , Línea Celular , Niño , Preescolar , Cristalografía por Rayos X , Citomegalovirus/efectos de los fármacos , Citomegalovirus/genética , Citomegalovirus/aislamiento & purificación , Infecciones por Citomegalovirus/diagnóstico , Infecciones por Citomegalovirus/etiología , Infecciones por Citomegalovirus/virología , ADN Viral/sangre , ADN Viral/aislamiento & purificación , Modelos Animales de Enfermedad , Farmacorresistencia Viral , Femenino , Ganciclovir/farmacología , Ganciclovir/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Interacciones Microbiota-Huesped/genética , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Muromegalovirus/aislamiento & purificación , Muromegalovirus/patogenicidad , Variantes Farmacogenómicas , Polimorfismo de Nucleótido Simple , Pirofosfatasas/metabolismo , Pirofosfatasas/ultraestructura , Resultado del Tratamiento , Adulto Joven
5.
Nature ; 593(7859): 362-371, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34012080

RESUMEN

Unlike the human genome that comprises mostly noncoding and regulatory sequences, viruses have evolved under the constraints of maintaining a small genome size while expanding the efficiency of their coding and regulatory sequences. As a result, viruses use strategies of transcription and translation in which one or more of the steps in the conventional gene-protein production line are altered. These alternative strategies of viral gene expression (also known as gene recoding) can be uniquely brought about by dedicated viral enzymes or by co-opting host factors (known as host dependencies). Targeting these unique enzymatic activities and host factors exposes vulnerabilities of a virus and provides a paradigm for the design of novel antiviral therapies. In this Review, we describe the types and mechanisms of unconventional gene and protein expression in viruses, and provide a perspective on how future basic mechanistic work could inform translational efforts that are aimed at viral eradication.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Regulación Viral de la Expresión Génica/efectos de los fármacos , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Virosis/tratamiento farmacológico , Virosis/virología , Animales , Sistema de Lectura Ribosómico/efectos de los fármacos , Sistema de Lectura Ribosómico/genética , Regulación Viral de la Expresión Génica/genética , Genoma Viral/efectos de los fármacos , Genoma Viral/genética , Humanos , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética
6.
IEEE/ACM Trans Comput Biol Bioinform ; 18(4): 1271-1280, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33891554

RESUMEN

COVID-19 is a highly contagious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The case-fatality rate is significantly higher in older patients and those with diabetes, cancer or cardiovascular disorders. The human proteins, angiotensin-converting enzyme 2 (ACE2), transmembrane protease serine 2 (TMPRSS2) and basigin (BSG), are involved in high-confidence host-pathogen interactions with SARS-CoV-2 proteins. We considered these three proteins as seed nodes and applied the random walk with restart method on the human interactome to construct a protein-protein interaction sub-network, which captures the effects of viral invasion. We found that 'Insulin resistance', 'AGE-RAGE signaling in diabetic complications' and 'adipocytokine signaling' were the common pathways associated with diabetes, cancer and cardiovascular disorders. The association of these critical pathways with aging and its related diseases explains the molecular basis of COVID-19 fatality. We further identified drugs that have effects on these proteins/pathways based on gene expression studies. We particularly focused on drugs that significantly downregulate ACE2 along with other critical proteins identified by the network-based approach. Among them, COL-3 had earlier shown activity against acute lung injury and acute respiratory distress, while entinostat and mocetinostat have been investigated for non-small-cell lung cancer. We propose that these drugs can be repurposed for COVID-19.


Asunto(s)
COVID-19/mortalidad , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/genética , Antivirales/uso terapéutico , COVID-19/epidemiología , COVID-19/terapia , Enfermedades Cardiovasculares/epidemiología , Comorbilidad , Biología Computacional , Reposicionamiento de Medicamentos , Enfermedades Gastrointestinales/epidemiología , Perfilación de la Expresión Génica/estadística & datos numéricos , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Pandemias , Mapas de Interacción de Proteínas/efectos de los fármacos , Enfermedades Respiratorias/epidemiología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Tratamiento Farmacológico de COVID-19
7.
Cancer Treat Res Commun ; 27: 100323, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33530025

RESUMEN

Human telomerase reverse transcriptase (hTERT) is an enzyme that is critically involved in elongating and maintaining telomeres length to control cell life span and replicative potential. Telomerase activity is continuously expressed in human germ-line cells and most cancer cells, whereas it is suppressed in most somatic cells. In normal cells, by reducing telomerase activity and progressively shortening the telomeres, the cells progress to the senescence or apoptosis process. However, in cancer cells, telomere lengths remain constant due to telomerase's reactivation, and cells continue to proliferate and inhibit apoptosis, and ultimately lead to cancer development and human death due to metastasis. Studies demonstrated that several DNA and RNA oncoviruses could interact with telomerase by integrating their genome sequence within the host cell telomeres specifically. Through the activation of the hTERT promoter and lengthening the telomere, these cells contributes to cancer development. Since oncoviruses can activate telomerase and increase hTERT expression, there are several therapeutic strategies based on targeting the telomerase of cancer cells like telomerase-targeted peptide vaccines, hTERT-targeting dendritic cells (DCs), hTERT-targeting gene therapy, and hTERT-targeting CRISPR/Cas9 system that can overcome tumor-mediated toleration mechanisms and specifically apoptosis in cancer cells. This study reviews available data on the molecular structure of telomerase and the role of oncoviruses and telomerase interaction in cancer development and telomerase-dependent therapeutic approaches to conquest the cancer cells.


Asunto(s)
Neoplasias/genética , Proteínas Oncogénicas Virales/metabolismo , Retroviridae/patogenicidad , Telomerasa/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/genética , Senescencia Celular/genética , Modelos Animales de Enfermedad , Terapia Genética/métodos , Interacciones Microbiota-Huesped/genética , Humanos , Ratones , Neoplasias/terapia , Neoplasias/virología , Proteínas Oncogénicas Virales/genética , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Regiones Promotoras Genéticas , Retroviridae/genética , Telomerasa/antagonistas & inhibidores , Telómero/metabolismo , Homeostasis del Telómero
8.
Sci Signal ; 14(665)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436497

RESUMEN

The first reported receptor for SARS-CoV-2 on host cells was the angiotensin-converting enzyme 2 (ACE2). However, the viral spike protein also has an RGD motif, suggesting that cell surface integrins may be co-receptors. We examined the sequences of ACE2 and integrins with the Eukaryotic Linear Motif (ELM) resource and identified candidate short linear motifs (SLiMs) in their short, unstructured, cytosolic tails with potential roles in endocytosis, membrane dynamics, autophagy, cytoskeleton, and cell signaling. These SLiM candidates are highly conserved in vertebrates and may interact with the µ2 subunit of the endocytosis-associated AP2 adaptor complex, as well as with various protein domains (namely, I-BAR, LC3, PDZ, PTB, and SH2) found in human signaling and regulatory proteins. Several motifs overlap in the tail sequences, suggesting that they may act as molecular switches, such as in response to tyrosine phosphorylation status. Candidate LC3-interacting region (LIR) motifs are present in the tails of integrin ß3 and ACE2, suggesting that these proteins could directly recruit autophagy components. Our findings identify several molecular links and testable hypotheses that could uncover mechanisms of SARS-CoV-2 attachment, entry, and replication against which it may be possible to develop host-directed therapies that dampen viral infection and disease progression. Several of these SLiMs have now been validated to mediate the predicted peptide interactions.


Asunto(s)
COVID-19/virología , Interacciones Microbiota-Huesped/fisiología , SARS-CoV-2/fisiología , SARS-CoV-2/patogenicidad , Internalización del Virus , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/fisiología , Animales , COVID-19/terapia , Secuencia Conservada , Interacciones Microbiota-Huesped/genética , Humanos , Integrinas/química , Integrinas/genética , Integrinas/fisiología , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/fisiología , Modelos Biológicos , Modelos Moleculares , Oligopéptidos/química , Oligopéptidos/genética , Oligopéptidos/fisiología , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/fisiología , Señales de Clasificación de Proteína/genética , Señales de Clasificación de Proteína/fisiología , Receptores Virales/química , Receptores Virales/genética , Receptores Virales/fisiología , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/fisiología
9.
Viruses ; 12(11)2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182673

RESUMEN

Dengue is an acute viral disease caused by Dengue virus (DENV) and is considered to be the most common arbovirus worldwide. The clinical characteristics of dengue may vary from asymptomatic to severe complications and severe organ impairment, particularly affecting the liver. Dengue treatment is palliative with acetaminophen (APAP), usually known as Paracetamol, being the most used drug aiming to relieve the mild symptoms of dengue. APAP is a safe and effective drug but, like dengue, can trigger the development of liver disorders. Given this scenario, it is necessary to investigate the effects of combining these two factors on hepatocyte homeostasis. Therefore, this study aimed to evaluate the molecular changes in hepatocytes resulting from the association between DENV infection and treatment with sub-toxic APAP concentrations. Using an in vitro experimental model of DENV-2 infected hepatocytes (AML-12 cells) treated with APAP, we evaluated the influence of the virus and drug association on the transcriptome of these hepatocytes by RNA sequencing (RNAseq). The virus-drug association was able to induce changes in the gene expression profile of AML-12 cells and here we highlight and explore these changes and its putative influence on biological processes for cellular homeostasis.


Asunto(s)
Acetaminofén/farmacología , Analgésicos no Narcóticos/farmacología , Virus del Dengue/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Interacciones Microbiota-Huesped , Transcriptoma , Animales , Línea Celular , Homeostasis/efectos de los fármacos , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Hígado/citología , Hígado/efectos de los fármacos , Hígado/virología , Ratones , Análisis de Secuencia de ARN , Replicación Viral/efectos de los fármacos
11.
Mol Pharm ; 17(10): 3649-3653, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32857512

RESUMEN

Adeno-associated virus (AAV)-based gene therapy is currently limited by (1) decline in therapeutic gene expression over time, (2) immune cell activation and (3) neutralization by pre-existing antibodies. Hence, studying the interaction of AAV vectors with various cellular pathways during the production and transduction process is necessary to overcome such barriers. Post-translational modifications (PTM) of AAV vectors during the production and transduction process is known to limit its transduction efficiency and further evoke the immune response. Further, AAV vectors are known to trigger cellular stress, resulting in an upregulation of distinct arms of the unfolded protein response (UPR) pathway. Recognition of the AAV genome by Toll-like receptor-9 triggers the myeloid differentiation primary response signaling cascade for innate (IL-6, IFN-α, IFN-ß) and adaptive (CD8+ T-cell, B-cell) immune response against the viral capsid and the transgene product. Herein, we highlight a potential intersection of the UPR, PTMs, and intracellular trafficking pathways, which could be fine-tuned to augment the outcome of AAV-based gene delivery.


Asunto(s)
Dependovirus/inmunología , Terapia Genética/métodos , Interacciones Microbiota-Huesped/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Transducción Genética/métodos , Inmunidad Adaptativa/genética , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Dependovirus/genética , Interacciones Microbiota-Huesped/genética , Humanos , Inmunidad Innata/genética , Procesamiento Proteico-Postraduccional/genética , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/inmunología
12.
Cell Host Microbe ; 28(3): 486-496.e6, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32738193

RESUMEN

There is an urgent need for vaccines and therapeutics to prevent and treat COVID-19. Rapid SARS-CoV-2 countermeasure development is contingent on the availability of robust, scalable, and readily deployable surrogate viral assays to screen antiviral humoral responses, define correlates of immune protection, and down-select candidate antivirals. Here, we generate a highly infectious recombinant vesicular stomatitis virus (VSV) bearing the SARS-CoV-2 spike glycoprotein S as its sole entry glycoprotein and show that this recombinant virus, rVSV-SARS-CoV-2 S, closely resembles SARS-CoV-2 in its entry-related properties. The neutralizing activities of a large panel of COVID-19 convalescent sera can be assessed in a high-throughput fluorescent reporter assay with rVSV-SARS-CoV-2 S, and neutralization of rVSV-SARS-CoV-2 S and authentic SARS-CoV-2 by spike-specific antibodies in these antisera is highly correlated. Our findings underscore the utility of rVSV-SARS-CoV-2 S for the development of spike-specific therapeutics and for mechanistic studies of viral entry and its inhibition.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/virología , Neumonía Viral/virología , Glicoproteína de la Espiga del Coronavirus/fisiología , Virus de la Estomatitis Vesicular Indiana/fisiología , Enzima Convertidora de Angiotensina 2 , Animales , Antivirales/farmacología , Betacoronavirus/genética , Betacoronavirus/fisiología , COVID-19 , Vacunas contra la COVID-19 , Línea Celular , Chlorocebus aethiops , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/terapia , Evaluación Preclínica de Medicamentos , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Mutación , Pruebas de Neutralización , Pandemias/prevención & control , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/fisiología , Neumonía Viral/prevención & control , Neumonía Viral/terapia , Receptores Virales/genética , Receptores Virales/fisiología , Recombinación Genética , SARS-CoV-2 , Serina Endopeptidasas/fisiología , Glicoproteína de la Espiga del Coronavirus/genética , Células Vero , Virus de la Estomatitis Vesicular Indiana/genética , Vacunas Virales/genética , Vacunas Virales/inmunología , Internalización del Virus , Replicación Viral/genética , Tratamiento Farmacológico de COVID-19
13.
Theranostics ; 10(16): 7034-7052, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32641977

RESUMEN

This review provides an update for the international research community on the cell modeling tools that could accelerate the understanding of SARS-CoV-2 infection mechanisms and could thus speed up the development of vaccines and therapeutic agents against COVID-19. Many bioengineering groups are actively developing frontier tools that are capable of providing realistic three-dimensional (3D) models for biological research, including cell culture scaffolds, microfluidic chambers for the culture of tissue equivalents and organoids, and implantable windows for intravital imaging. Here, we review the most innovative study models based on these bioengineering tools in the context of virology and vaccinology. To make it easier for scientists working on SARS-CoV-2 to identify and apply specific tools, we discuss how they could accelerate the discovery and preclinical development of antiviral drugs and vaccines, compared to conventional models.


Asunto(s)
Antivirales/aislamiento & purificación , Antivirales/farmacología , Betacoronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/prevención & control , Vacunas Virales/aislamiento & purificación , Vacunas Virales/farmacología , Betacoronavirus/química , Betacoronavirus/genética , Betacoronavirus/inmunología , Bioingeniería/métodos , Bioingeniería/tendencias , Reactores Biológicos , COVID-19 , Vacunas contra la COVID-19 , Técnicas de Cultivo de Célula , Simulación por Computador , Infecciones por Coronavirus/inmunología , Descubrimiento de Drogas/métodos , Descubrimiento de Drogas/tendencias , Evaluación de Medicamentos/métodos , Evaluación de Medicamentos/tendencias , Farmacorresistencia Viral , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Modelos Biológicos , Organoides/citología , Organoides/virología , Neumonía Viral/inmunología , SARS-CoV-2 , Nanomedicina Teranóstica
14.
Comput Biol Med ; 121: 103749, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32568687

RESUMEN

This paper continues a recent study of the spike protein sequence of the COVID-19 virus (SARS-CoV-2). It is also in part an introductory review to relevant computational techniques for tackling viral threats, using COVID-19 as an example. Q-UEL tools for facilitating access to knowledge and bioinformatics tools were again used for efficiency, but the focus in this paper is even more on the virus. Subsequence KRSFIEDLLFNKV of the S2' spike glycoprotein proteolytic cleavage site continues to appear important. Here it is shown to be recognizable in the common cold coronaviruses, avian coronaviruses and possibly as traces in the nidoviruses of reptiles and fish. Its function or functions thus seem important to the coronaviruses. It might represent SARS-CoV-2 Achilles' heel, less likely to acquire resistance by mutation, as has happened in some early SARS vaccine studies discussed in the previous paper. Preliminary conformational analysis of the receptor (ACE2) binding site of the spike protein is carried out suggesting that while it is somewhat conserved, it appears to be more variable than KRSFIEDLLFNKV. However compounds like emodin that inhibit SARS entry, apparently by binding ACE2, might also have functions at several different human protein binding sites. The enzyme 11ß-hydroxysteroid dehydrogenase type 1 is again argued to be a convenient model pharmacophore perhaps representing an ensemble of targets, and it is noted that it occurs both in lung and alimentary tract. Perhaps it benefits the virus to block an inflammatory response by inhibiting the dehydrogenase, but a fairly complex web involves several possible targets.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/prevención & control , Glicoproteína de la Espiga del Coronavirus/química , Vacunas Virales/inmunología , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2 , Animales , Antivirales/farmacología , Betacoronavirus/química , Betacoronavirus/genética , Betacoronavirus/inmunología , Sitios de Unión , COVID-19 , Vacunas contra la COVID-19 , Biología Computacional , Coronavirus/química , Coronavirus/genética , Coronavirus/inmunología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Diseño de Fármacos , Farmacorresistencia Viral/genética , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Modelos Moleculares , Mutación , Peptidomiméticos/farmacología , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/virología , SARS-CoV-2 , Homología de Secuencia de Aminoácido , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/genética
15.
Microbiome ; 8(1): 53, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32299497

RESUMEN

BACKGROUND: Recent evidence has linked the gut microbiome to host behavior via the gut-brain axis [1-3]; however, the underlying mechanisms remain unexplored. Here, we determined the links between host genetics, the gut microbiome and memory using the genetically defined Collaborative Cross (CC) mouse cohort, complemented with microbiome and metabolomic analyses in conventional and germ-free (GF) mice. RESULTS: A genome-wide association analysis (GWAS) identified 715 of 76,080 single-nucleotide polymorphisms (SNPs) that were significantly associated with short-term memory using the passive avoidance model. The identified SNPs were enriched in genes known to be involved in learning and memory functions. By 16S rRNA gene sequencing of the gut microbial community in the same CC cohort, we identified specific microorganisms that were significantly correlated with longer latencies in our retention test, including a positive correlation with Lactobacillus. Inoculation of GF mice with individual species of Lactobacillus (L. reuteri F275, L. plantarum BDGP2 or L. brevis BDGP6) resulted in significantly improved memory compared to uninoculated or E. coli DH10B inoculated controls. Untargeted metabolomics analysis revealed significantly higher levels of several metabolites, including lactate, in the stools of Lactobacillus-colonized mice, when compared to GF control mice. Moreover, we demonstrate that dietary lactate treatment alone boosted memory in conventional mice. Mechanistically, we show that both inoculation with Lactobacillus or lactate treatment significantly increased the levels of the neurotransmitter, gamma-aminobutyric acid (GABA), in the hippocampus of the mice. CONCLUSION: Together, this study provides new evidence for a link between Lactobacillus and memory and our results open possible new avenues for treating memory impairment disorders using specific gut microbial inoculants and/or metabolites. Video Abstract.


Asunto(s)
Bacterias/clasificación , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped/genética , Memoria , Animales , Suplementos Dietéticos , Heces/química , Femenino , Estudio de Asociación del Genoma Completo , Vida Libre de Gérmenes , Lactatos/administración & dosificación , Lactobacillus , Masculino , Metabolómica , Ratones/genética , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple , ARN Ribosómico 16S , Ácido gamma-Aminobutírico/análisis
16.
Dev Comp Immunol ; 109: 103717, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32348787

RESUMEN

Corals are comprised of a coral host and associated microbes whose interactions are mediated by the coral innate immune system. The diversity of immune factors identified in the Pocillopora damicornis genome suggests that immunity is linked to maintaining microbial symbioses while also being able to detect pathogens. However, it is unclear which immune factors respond to specific microbe-associated molecular patterns and how these immune reactions simultaneously affect coral-associated bacteria. To investigate this, fragments of P. damicornis and P. acuta colonies from Taiwan were subjected to lipopolysaccharide (LPS) treatment to stimulate immune responses and measure bacteria community shifts. RNA-seq revealed genotype-specific immune responses to LPS involving the upregulation of immune receptors, transcription factors, and pore-forming toxins. Bacteria 16S sequencing revealed significantly different bacteria communities between coral genotypes but no differences in bacteria communities were caused by LPS. Our findings confirm that Pocillopora corals activate conserved immune factors in response to LPS and identify transcription factors coordinating Pocillopora corals' immune responses. Additionally, the strong effect of coral genotype on gene expression and bacteria communities highlights the importance of coral genotype in the investigation of coral host-microbe interactions.


Asunto(s)
Antozoos/inmunología , Arrecifes de Coral , Inmunidad/efectos de los fármacos , Lipopolisacáridos/farmacología , Animales , Antozoos/genética , Antozoos/microbiología , Bacterias/clasificación , Bacterias/genética , Ecosistema , Regulación de la Expresión Génica/efectos de los fármacos , Ontología de Genes , Genotipo , Interacciones Microbiota-Huesped/genética , Inmunidad/genética , ARN Ribosómico 16S/genética
17.
Viruses ; 12(1)2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31936258

RESUMEN

Beet necrotic yellow vein virus (BNYVV) and Beet soil-borne mosaic virus (BSBMV) are closely related species, but disease development induced in their host sugar beet displays striking differences. Beet necrotic yellow vein virus induces excessive lateral root (LR) formation, whereas BSBMV-infected roots appear asymptomatic. A comparative transcriptome analysis was performed to elucidate transcriptomic changes associated with disease development. Many differentially expressed genes (DEGs) were specific either to BNYVV or BSBMV, although both viruses shared a high number of DEGs. Auxin biosynthesis pathways displayed a stronger activation by BNYVV compared to BSBMV-infected plants. Several genes regulated by auxin signalling and required for LR formation were exclusively altered by BNYVV. Both viruses reprogrammed the transcriptional network, but a large number of transcription factors involved in plant defence were upregulated in BNYVV-infected plants. A strong activation of pathogenesis-related proteins by both viruses suggests a salicylic acid or jasmonic acid mediated-defence response, but the data also indicate that both viruses counteract the SA-mediated defence. The ethylene signal transduction pathway was strongly downregulated which probably increases the susceptibility of sugar beet to Benyvirus infection. Our study provides a deeper insight into the interaction of BNYVV and BSBMV with the economically important crop sugar beet.


Asunto(s)
Beta vulgaris/virología , Interacciones Microbiota-Huesped/genética , Virus del Mosaico/genética , Virus de Plantas/genética , Microbiología del Suelo , Vías Biosintéticas , Perfilación de la Expresión Génica , Enfermedades de las Plantas/virología , Raíces de Plantas/fisiología , Raíces de Plantas/virología , Transducción de Señal
18.
PLoS One ; 14(11): e0216184, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31693670

RESUMEN

The increased recurrence of Candida albicans infections is associated with greater resistance to antifungal drugs. This involves the establishment of alternative therapeutic protocols, such as probiotic microorganisms whose antifungal potential has already been demonstrated using preclinical models (cell cultures, laboratory animals). Understanding the mechanisms of action of probiotic microorganisms has become a strategic need for the development of new therapeutics for humans. In this study, we investigated the prophylactic anti-C. albicans properties of Lactobacillus rhamnosus Lcr35® using the in vitro Caco-2 cell model and the in vivo Caenorhabditis elegans model. In Caco-2 cells, we showed that the strain Lcr35® significantly inhibited the growth (~2 log CFU.mL-1) and adhesion (150 to 6,300 times less) of the pathogen. Moreover, in addition to having a pro-longevity activity in the nematode (+42.9%, p = 3.56.10-6), Lcr35® protects the animal from the fungal infection (+267% of survival, p < 2.10-16) even if the yeast is still detectable in its intestine. At the mechanistic level, we noticed the repression of genes of the p38 MAPK signalling pathway and genes involved in the antifungal response induced by Lcr35®, suggesting that the pathogen no longer appears to be detected by the worm immune system. However, the DAF-16/FOXO transcription factor, implicated in the longevity and antipathogenic response of C. elegans, is activated by Lcr35®. These results suggest that the probiotic strain acts by stimulating its host via DAF-16 but also by suppressing the virulence of the pathogen.


Asunto(s)
Candida albicans , Candidiasis/prevención & control , Lacticaseibacillus rhamnosus , Probióticos/uso terapéutico , Transporte Activo de Núcleo Celular , Animales , Animales Modificados Genéticamente , Células CACO-2 , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiología , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Candida albicans/crecimiento & desarrollo , Candida albicans/patogenicidad , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Virulencia
19.
Artículo en Inglés | MEDLINE | ID: mdl-31475121

RESUMEN

Ticks transmit the most diverse array of disease agents and harbor one of the most diverse microbial communities. Major progress has been made in the characterization of the taxonomic profiles of tick microbiota. However, the functional profiles of tick microbiome have been comparatively less studied. In this proof of concept we used state-of-the-art functional metagenomics analytical tools to explore previously reported datasets of bacteria found in male and female Ixodes ovatus, Ixodes persulcatus, and Amblyomma variegatum. Results showed that both taxonomic and functional profiles have differences between sexes of the same species. KEGG pathway analysis revealed that male and female of the same species had major differences in the abundance of genes involved in different metabolic pathways including vitamin B, amino acids, carbohydrates, nucleotides, and antibiotics among others. Partial reconstruction of metabolic pathways using KEGG enzymes suggests that tick microbiome form a complex metabolic network that may increase microbial community resilience and adaptability. Linkage analysis between taxonomic and functional profiles showed that among the KEGG enzymes with differential abundance in male and female ticks only 12% were present in single bacterial genera. The rest of these enzymes were found in more than two bacterial genera, and 27% of them were found in five up to ten bacterial genera. Comparison of bacterial genera contributing to the differences in the taxonomic and functional profiles of males and females revealed that while a small group of bacteria has a dual-role, most of the bacteria contribute only to functional or taxonomic differentiation between sexes. Results suggest that the different life styles of male and female ticks exert sex-specific evolutionary pressures that act independently on the phenomes (set of phenotypes) and genomes of bacteria in tick gut microbiota. We conclude that functional redundancy is a fundamental property of male and female tick microbiota and propose that functional metagenomics should be combined with taxonomic profiling of microbiota because both analyses are complementary.


Asunto(s)
Microbioma Gastrointestinal , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Garrapatas/genética , Garrapatas/metabolismo , Garrapatas/microbiología , Aminoácidos/metabolismo , Animales , Bacterias/clasificación , Bacterias/genética , Metabolismo de los Hidratos de Carbono/genética , Enzimas/genética , Enzimas/metabolismo , Femenino , Microbioma Gastrointestinal/genética , Ixodes/microbiología , Masculino , Redes y Vías Metabólicas/genética , Metagenómica/métodos , Filogenia , ARN Ribosómico 16S/genética , Factores Sexuales
20.
Curr Drug Targets ; 20(16): 1636-1651, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31362671

RESUMEN

BACKGROUND: Hepatitis B is a very harmful and epidemic disease caused by hepatitis B virus (HBV). Although an effective anti-HBV vaccine is available, chronic infection poses still a huge health burden in the whole world. The present anti-HBV drugs including nucleoside analogues and interferonalpha have their limitations without exception. There is no effective drug and therapeutic method that can really and truly cure hepatitis B so far. The variability of HBV genome results in that a significant number of patients develop drug resistance during the long-term use of anti-HBV drugs. Hence, it is urgently needed to discover novel targets and develop new drugs against hepatitis B. OBJECTIVE: The review aims to provide the theory support for designing of the anti-HBV innovative drugs by offering a summary of the current situation of antiviral potential targets. RESULTS AND CONCLUSION: Since HBV is obligate intracellular parasite, and as such it depends on host cellular components and functions to replicate itself. The targeting both virus and host might be a novel therapeutic option for hepatitis B. Accordingly, we analyse the advances in the study of the potential drug targets for anti-HBV infection, focusing on targeting virus genome, on targeting host cellular functions and on targeting virus-host proteins interactions, respectively. Meanwhile, the immune targets against chronic hepatitis B are also emphasized. In short, the review provides a summary of antiviral therapeutic strategies to target virus factors, host factors and immune factors for future designing of the innovative drug against HBV infection.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B Crónica/tratamiento farmacológico , Animales , Descubrimiento de Drogas/métodos , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Genoma Viral/efectos de los fármacos , Genoma Viral/genética , Virus de la Hepatitis B/genética , Hepatitis B Crónica/virología , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA