Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(10): e1011682, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37782657

RESUMEN

Human cytomegalovirus (HCMV) encodes multiple putative G protein-coupled receptors (GPCRs). US28 functions as a viral chemokine receptor and is expressed during both latent and lytic phases of virus infection. US28 actively promotes cellular migration, transformation, and plays a major role in mediating viral latency and reactivation; however, knowledge about the interaction partners involved in these processes is still incomplete. Herein, we utilized a proximity-dependent biotinylating enzyme (TurboID) to characterize the US28 interactome when expressed in isolation, and during both latent (CD34+ hematopoietic progenitor cells) and lytic (fibroblasts) HCMV infection. Our analyses indicate that the US28 signalosome converges with RhoA and EGFR signal transduction pathways, sharing multiple mediators that are major actors in processes such as cellular proliferation and differentiation. Integral members of the US28 signaling complex were validated in functional assays by immunoblot and small-molecule inhibitors. Importantly, we identified RhoGEFs as key US28 signaling intermediaries. In vitro latency and reactivation assays utilizing primary CD34+ hematopoietic progenitor cells (HPCs) treated with the small-molecule inhibitors Rhosin or Y16 indicated that US28 -RhoGEF interactions are required for efficient viral reactivation. These findings were recapitulated in vivo using a humanized mouse model where inhibition of RhoGEFs resulted in a failure of the virus to reactivate. Together, our data identifies multiple new proteins in the US28 interactome that play major roles in viral latency and reactivation, highlights the utility of proximity-sensor labeling to characterize protein interactomes, and provides insight into targets for the development of novel anti-HCMV therapeutics.


Asunto(s)
Citomegalovirus , Transducción de Señal , Animales , Ratones , Humanos , Citomegalovirus/fisiología , Latencia del Virus , Diferenciación Celular , Células Madre Hematopoyéticas
2.
Antimicrob Agents Chemother ; 65(9): e0024421, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34152810

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a reemerging alphavirus that can cause encephalitis resulting in severe human morbidity and mortality. Using a high-throughput cell-based screen, we identified a quinolinone compound that protected against VEEV-induced cytopathic effects. Analysis of viral replication in cells identified several quinolinone compounds with potent inhibitory activity against vaccine and virulent strains of VEEV. These quinolinones also displayed inhibitory activity against additional alphaviruses, such as Mayaro virus and Ross River virus, although the potency was greatly reduced. Time-of-addition studies indicated that these compounds inhibit the early-to-mid stage of viral replication. Deep sequencing and reverse genetics studies identified two unique resistance mutations in the nsP2 gene (Y102S/C; stalk domain) that conferred VEEV resistance on this chemical series. Moreover, introduction of a K102Y mutation into the nsP2 gene enhanced the sensitivity of chikungunya virus (CHIKV) to this chemical series. Computational modeling of CHIKV and VEEV nsP2 identified a highly probable docking alignment for the quinolinone compounds that require a tyrosine residue at position 102 within the helicase stalk domain. These studies identified a class of compounds with antiviral activity against VEEV and other alphaviruses and provide further evidence that therapeutics targeting nsP2 may be useful against alphavirus infection.


Asunto(s)
Virus Chikungunya , Virus de la Encefalitis Equina Venezolana , Quinolonas , Animales , Antivirales/farmacología , Virus de la Encefalitis Equina Venezolana/genética , Caballos , Humanos , Quinolonas/farmacología , Replicación Viral
4.
PLoS Pathog ; 13(3): e1006219, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28278237

RESUMEN

Zika virus (ZIKV), an emerging flavivirus, has recently spread explosively through the Western hemisphere. In addition to symptoms including fever, rash, arthralgia, and conjunctivitis, ZIKV infection of pregnant women can cause microcephaly and other developmental abnormalities in the fetus. We report herein the results of ZIKV infection of adult rhesus macaques. Following subcutaneous infection, animals developed transient plasma viremia and viruria from 1-7 days post infection (dpi) that was accompanied by the development of a rash, fever and conjunctivitis. Animals produced a robust adaptive immune response to ZIKV, although systemic cytokine response was minimal. At 7 dpi, virus was detected in peripheral nervous tissue, multiple lymphoid tissues, joints, and the uterus of the necropsied animals. Notably, viral RNA persisted in neuronal, lymphoid and joint/muscle tissues and the male and female reproductive tissues through 28 to 35 dpi. The tropism and persistence of ZIKV in the peripheral nerves and reproductive tract may provide a mechanism of subsequent neuropathogenesis and sexual transmission.


Asunto(s)
Infección por el Virus Zika/patología , Infección por el Virus Zika/virología , Animales , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Hibridación in Situ , Macaca mulatta , Masculino , Pruebas de Neutralización , Reacción en Cadena de la Polimerasa , Viremia/virología , Virus Zika
5.
Int J Hyperthermia ; 36(1): 466-472, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30922131

RESUMEN

Objective/Purpose: In order to study the effects of hyperthermia and other temperature-related effects on cells and tissues, determining the precise time/temperature course is crucial. Here we present a non-contact optoacoustic technique, which provides temperatures during heating of cultured cells with scalable temporal and spatial resolution. METHODS: A thulium laser (1.94 µm) with a maximum power of 15 W quickly and efficiently heats cells in a culture dish because of low penetration depth (1/e penetration depths of 78 µm) of the radiation in water. A repetitively Q-switched holmium laser (2.1 µm) is used simultaneously to probe temperatures at different locations in the dish by using the photoacoustic effect. Due to thermoelastic expansion of water, pressure waves are emitted and measured with an ultrasonic hydrophone at the side of the dish. The amplitudes of the waves are temperature dependent and can be used to calculate the temperature/time course at any location of probing. RESULTS: We measured temperatures of up to 55 °C with a heating power of 6 W after 10 s, and subsequent lateral temperature profiles over time. Within this profile, temperature fluctuations were found, likely owing to thermal convection and water circulation. By using cultured retinal pigment epithelial cells, it is shown that the probe laser pulses alone cause no biological damage, while immediate cell damage occurs when heating for 10 s at temperatures exceeding 45 °C. CONCLUSIONS: This method shows great potential not only as a noninvasive, non-contact method to determine temperature/time responses of cells in culture, but also for complex tissue and other materials.


Asunto(s)
Calor/uso terapéutico , Hipertermia Inducida/métodos , Células Cultivadas , Estudios de Factibilidad , Humanos
6.
J Virol ; 88(15): 8213-26, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24829346

RESUMEN

UNLABELLED: Chikungunya virus (CHIKV) is a reemerging mosquito-transmitted alphavirus that causes epidemics of debilitating polyarthritis in humans. A prior study identified two anti-CHIKV monoclonal antibodies ([MAbs] CHK-152 and CHK-166) against the E2 and E1 structural proteins, which had therapeutic efficacy in immunocompetent and immunocompromised mice. Combination MAb therapy was required as administration of a single MAb resulted in the rapid selection of neutralization escape variants and treatment failure in mice. Here, we initially evaluated the efficacy of combination MAb therapy in a nonhuman primate model of CHIKV infection. Treatment of rhesus macaques with CHK-152 and CHK-166 reduced viral spread and infection in distant tissue sites and also neutralized reservoirs of infectious virus. Escape viruses were not detected in the residual viral RNA present in tissues and organs of rhesus macaques. To evaluate the possible significance of MAb resistance, we engineered neutralization escape variant viruses (E1-K61T, E2-D59N, and the double mutant E1-K61T E2-D59N) that conferred resistance to CHK-152 and CHK-166 and tested them for fitness in mosquito cells, mammalian cells, mice, and Aedes albopictus mosquitoes. In both cell culture and mosquitoes, the mutant viruses grew equivalently and did not revert to wild-type (WT) sequence. All escape variants showed evidence of mild clinical attenuation, with decreased musculoskeletal disease at early times after infection in WT mice and a prolonged survival time in immunocompromised Ifnar1(-/-) mice. Unexpectedly, this was not associated with decreased infectivity, and consensus sequencing from tissues revealed no evidence of reversion or compensatory mutations. Competition studies with CHIKV WT also revealed no fitness compromise of the double mutant (E1-K61T E2-D59N) neutralization escape variant in WT mice. Collectively, our study suggests that neutralization escape viruses selected during combination MAb therapy with CHK-152 plus CHK-166 retain fitness, cause less severe clinical disease, and likely would not be purified during the enzootic cycle. IMPORTANCE: Chikungunya virus (CHIKV) causes explosive epidemics of acute and chronic arthritis in humans in Africa, the Indian subcontinent, and Southeast Asia and recently has spread to the New World. As there are no approved vaccines or therapies for human use, the possibility of CHIKV-induced debilitating disease is high in many parts of the world. To this end, our laboratory recently generated a combination monoclonal antibody therapy that aborted lethal and arthritogenic disease in wild-type and immunocompromised mice when administered as a single dose several days after infection. In this study, we show the efficacy of the antibody combination in nonhuman primates and also evaluate the significance of possible neutralization escape mutations in mosquito and mammalian cells, mice, and Aedes albopictus vector mosquitoes. Our experiments show that escape viruses from combination antibody therapy cause less severe CHIKV clinical disease, retain fitness, and likely would not be purified by mosquito vectors.


Asunto(s)
Aedes/virología , Infecciones por Alphavirus/virología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Virus Chikungunya/fisiología , Inmunoterapia/métodos , Replicación Viral , Infecciones por Alphavirus/terapia , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Fiebre Chikungunya , Virus Chikungunya/inmunología , Virus Chikungunya/aislamiento & purificación , Virus Chikungunya/patogenicidad , Modelos Animales de Enfermedad , Femenino , Proteínas de Homeodominio , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia , Virulencia
7.
bioRxiv ; 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38464092

RESUMEN

Human cytomegalovirus (HCMV) encodes four viral Fc-gamma receptors (vFcγRs) that counteract antibody-mediated activation in vitro , but their role in infection and pathogenesis is unknown. To examine the in vivo function of vFcγRs in animal hosts closely related to humans, we identified and characterized vFcγRs encoded by rhesus CMV (RhCMV). We demonstrate that Rh05, Rh152/151 and Rh173 represent the complete set of RhCMV vFcγRs, each displaying functional similarities to their respective HCMV orthologs with respect to antagonizing host FcγR activation in vitro . When RhCMV-naïve rhesus macaques were infected with vFcγR-deleted RhCMV, peak plasma viremia levels and anti-RhCMV antibody responses were comparable to wildtype infections. However, the duration of plasma viremia was significantly shortened in immunocompetent, but not in CD4+ T cell-depleted animals. Since vFcγRs were not required for superinfection, we conclude that vFcγRs delay control by virus-specific adaptive immune responses, particularly antibodies, during primary infection.

8.
J Virol ; 86(21): 11833-44, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22915808

RESUMEN

Cytomegaloviruses manipulate the host chemokine/receptor axis by altering cellular chemokine expression and by encoding multiple chemokines and chemokine receptors. Similar to human cytomegalovirus (HCMV), rat cytomegalovirus (RCMV) encodes multiple CC chemokine-analogous proteins, including r129 (HCMV UL128 homologue) and r131 (HCMV UL130 and MCMV m129/130 homologues). Although these proteins play a role in CMV entry, their function as chemotactic cytokines remains unknown. In the current study, we examined the role of the RCMV chemokine r129 in promoting cellular migration and in accelerating transplant vascular sclerosis (TVS) in our rat heart transplant model. We determined that r129 protein is released into culture supernatants of infected cells and is expressed with late viral gene kinetics during RCMV infection and highly expressed in heart and salivary glands during in vivo rat infections. Using the recombinant r129 protein, we demonstrated that r129 induces migration of lymphocytes isolated from rat peripheral blood, spleen, and bone marrow and from a rat macrophage cell line. Using antibody-mediated cell sorting of rat splenocytes, we demonstrated that r129 induces migration of naïve/central memory CD4(+) T cells. Through ligand-binding assays, we determined that r129 binds rat CC chemokine receptors CCR3, CCR4, CCR5, and CCR7. In addition, mutational analyses identified functional domains of r129 resulting in recombinant proteins that fail to induce migration (r129-ΔNT and -C31A) or alter the chemotactic ability of the chemokine (r129-F43A). Two of the mutant proteins (r129-C31A and -ΔNT) also act as dominant negatives by inhibiting migration induced by wild-type r129. Furthermore, infection of rat heart transplant recipients with RCMV containing the r129-ΔNT mutation prevented CMV-induced acceleration of TVS. Together our findings indicate that RCMV r129 is highly chemotactic, which has important implications during RCMV infection and reactivation and acceleration of TVS.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Movimiento Celular , Quimiocinas CC/inmunología , Muromegalovirus/inmunología , Muromegalovirus/patogenicidad , Factores de Virulencia/inmunología , Factores de Virulencia/metabolismo , Animales , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Trasplante de Corazón/efectos adversos , Infecciones por Herpesviridae/complicaciones , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Masculino , Muromegalovirus/genética , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Proteínas Mutantes/metabolismo , Unión Proteica , Ratas , Receptores CCR/metabolismo , Esclerosis/inmunología , Esclerosis/patología , Esclerosis/virología , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/patología , Enfermedades Vasculares/virología , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Factores de Virulencia/genética
9.
Proc Natl Acad Sci U S A ; 107(47): 20335-9, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21059930

RESUMEN

Achieving label-free, molecular-specific imaging with high spatial resolution in deep tissue is often considered the grand challenge of optical imaging. To accomplish this goal, significant optical scattering in tissues has to be overcome while achieving molecular specificity without resorting to extrinsic labeling. We demonstrate the feasibility of developing such an optical imaging modality by combining the molecularly specific stimulated Raman excitation with the photoacoustic detection. By employing two ultrashort excitation laser pulses, separated in frequency by the vibrational frequency of a targeted molecule, only the specific vibrational level of the target molecules in the illuminated tissue volume is excited. This targeted optical absorption generates ultrasonic waves (referred to as stimulated Raman photoacoustic waves) which are detected using a traditional ultrasonic transducer to form an image following the design of the established photoacoustic microscopy.


Asunto(s)
Rayos Láser , Imagen Molecular/métodos , Espectrometría Raman , Ultrasonido/métodos , Cloroformo/química , Modelos Teóricos , Vibración
10.
Mitochondrion ; 68: 15-24, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36371074

RESUMEN

Photobiomodulation is a term for using low-power red to near-infrared light to stimulate a variety of positive biological effects. Though the scientific and clinical acceptance of PBM as a therapeutic intervention has increased dramatically in recent years, the molecular underpinnings of the effect remain poorly understood. The putative chromophore for PBM effects is cytochrome c oxidase. It is postulated that light absorption at cytochrome c oxidase initiates a signaling cascade involving ATP and generation of reactive oxygen species (ROS), which subsequently results in improved cellular robustness. However, this hypothesis is largely based on inference and indirect evidence, and the precise molecular mechanisms that govern how photon absorption leads to these downstream effects remain poorly understood. We conducted low-power PBM-type light exposures of isolated mitochondria to 808 nm NIR light, at a number of irradiances. NIR exposure was found to enhance the activity of complex IV, depress the activity of complex III, and had no effect on the activity of complex II. Further, examining the dose-response of complex IV we found NIR enhancement did not exhibit irradiance reciprocity, indicating the effect on complex IV may not have direct photochemical basis. In summary, this research presents a novel method to interrogate the earliest stages of PBM in the mitochondria, and a unique window into the corresponding molecular mechanism(s) of induction.


Asunto(s)
Complejo IV de Transporte de Electrones , Terapia por Luz de Baja Intensidad , Complejo IV de Transporte de Electrones/metabolismo , Transporte de Electrón , Terapia por Luz de Baja Intensidad/métodos , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo
11.
PLoS Negl Trop Dis ; 17(3): e0011154, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36913428

RESUMEN

Infections with Chikungunya virus, a mosquito-borne alphavirus, cause an acute febrile syndrome often followed by chronic arthritis that persists for months to years post-infection. Neutralizing antibodies are the primary immune correlate of protection elicited by infection, and the major goal of vaccinations in development. Using convalescent blood samples collected from both endemic and non-endemic human subjects at multiple timepoints following suspected or confirmed chikungunya infection, we identified antibodies with broad neutralizing properties against other alphaviruses within the Semliki Forest complex. Cross-neutralization generally did not extend to the Venezuelan Equine Encephalitis virus (VEEV) complex, although some subjects had low levels of VEEV-neutralizing antibodies. This suggests that broadly neutralizing antibodies elicited following natural infection are largely complex restricted. In addition to serology, we also performed memory B-cell analysis, finding chikungunya-specific memory B-cells in all subjects in this study as remotely as 24 years post-infection. We functionally assessed the ability of memory B-cell derived antibodies to bind to chikungunya virus, and related Mayaro virus, as well as the highly conserved B domain of the E2 glycoprotein thought to contribute to cross-reactivity between related Old-World alphaviruses. To specifically assess the role of the E2 B domain in cross-neutralization, we depleted Mayaro and Chikungunya virus E2 B domain specific antibodies from convalescent sera, finding E2B depletion significantly decreases Mayaro virus specific cross-neutralizing antibody titers with no significant effect on chikungunya virus neutralization, indicating that the E2 B domain is a key target of cross-neutralizing and potentially cross-protective neutralizing antibodies.


Asunto(s)
Fiebre Chikungunya , Virus Chikungunya , Animales , Humanos , Anticuerpos ampliamente neutralizantes , Anticuerpos Antivirales , Anticuerpos Neutralizantes , Glicoproteínas
12.
PLoS Negl Trop Dis ; 17(11): e0011742, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37983245

RESUMEN

Mayaro virus (MAYV) is a mosquito-transmitted alphavirus that causes debilitating and persistent arthritogenic disease. While MAYV was previously reported to infect non-human primates (NHP), characterization of MAYV pathogenesis is currently lacking. Therefore, in this study we characterized MAYV infection and immunity in rhesus macaques. To inform the selection of a viral strain for NHP experiments, we evaluated five MAYV strains in C57BL/6 mice and showed that MAYV strain BeAr505411 induced robust tissue dissemination and disease. Three male rhesus macaques were subcutaneously challenged with 105 plaque-forming units of this strain into the arms. Peak plasma viremia occurred at 2 days post-infection (dpi). NHPs were taken to necropsy at 10 dpi to assess viral dissemination, which included the muscles and joints, lymphoid tissues, major organs, male reproductive tissues, as well as peripheral and central nervous system tissues. Histological examination demonstrated that MAYV infection was associated with appendicular joint and muscle inflammation as well as presence of perivascular inflammation in a wide variety of tissues. One animal developed a maculopapular rash and two NHP had viral RNA detected in upper torso skin samples, which was associated with the presence of perivascular and perifollicular lymphocytic aggregation. Analysis of longitudinal peripheral blood samples indicated a robust innate and adaptive immune activation, including the presence of anti-MAYV neutralizing antibodies with activity against related Una virus and chikungunya virus. Inflammatory cytokines and monocyte activation also peaked coincident with viremia, which was well supported by our transcriptomic analysis highlighting enrichment of interferon signaling and other antiviral processes at 2 days post MAYV infection. The rhesus macaque model of MAYV infection recapitulates many of the aspects of human infection and is poised to facilitate the evaluation of novel therapies and vaccines targeting this re-emerging virus.


Asunto(s)
Infecciones por Alphavirus , Alphavirus , Virus Chikungunya , Animales , Ratones , Masculino , Macaca mulatta , Viremia , Ratones Endogámicos C57BL , Anticuerpos Antivirales
13.
Aust J Chem ; 65(3): 260-265, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23807817

RESUMEN

A powerful combination of chemically specific Raman excitation and deep tissue ultrasound imaging holds the promise to attain spatially resolved distribution of chemical compounds inside the scattering medium. In this report, an attempt is made to evaluate the recent achievements and possible challenges with an eye on potential clinical applications.

14.
J Biomed Opt ; 27(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35362274

RESUMEN

SIGNIFICANCE: Physics-based models supply simulated temperature rises to photothermal damage rate models and provide comprehensive risk assessments for laser-induced damage. As the physics-based models continue to be refined, the damage rate models have not advanced. This peculiar lack of improvement is counterintuitive considering the damage integral (Ω), originally derived for isothermal heating events, and fails to accurately represent the nonisothermal heating from short laser exposures. AIM: Derive a nonisothermal form of the damage integral and predict more accurately the damage induced by short laser exposures, as well as identify the role of heating rate in laser damage. APPROACH: From first principles, we derived a version of the damage integral specific to the shape of thermal profiles rather than the square function provided by Arrhenius plots. We used previously published threshold thermal profiles, where all nonisothermal frequency factors (Anon) solved all Ωnon values to unity. Nonisothermal correction factors correct isothermal Aiso values. RESULTS: The Ea values were identical for both the isothermal and nonisothermal conventions. Correction factor values for Ωiso ranged from 0.0 (20-s exposures at thermal steady state) to -0.93 (0.05-s exposures). Based on empirical results, we have derived a two-dimensional empirical formula that predicts the heating rate as a function of exposure duration and ambient temperature. Threshold peak temperatures (Tpthr) and threshold critical temperatures are mathematically determined without thermal profiles when appropriate Ea and Anon values are established. CONCLUSIONS: We have identified a modified damage integral that does not rely on the Arrhenius plot and provides a value for the frequency factor (A) that accounts for the nonisothermal nature of short laser exposures. The method, validated in our in vitro retinal model, requires thermal profiles recorded under threshold conditions, such as at minimum visible lesions or the boundary of cell death. The method is a new option for laser damage modelers.


Asunto(s)
Calefacción , Rayos Láser , Retina , Temperatura
15.
Opt Lett ; 36(7): 1233-5, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21479040

RESUMEN

A capability of high-frequency ultrasound detection to monitor the process of energy deposition into a molecular system via Raman excitation is experimentally demonstrated. It is shown that the generated ultrasound signal is directly proportional to the optical signal generated in stimulated Raman scattering. Ultrasound detection provides a simple way to discriminate against laser-induced breakdown and allows for the quantification of the stimulated Raman scattering process where direct optical detection is not available. Additionally, it can be used for stimulated Raman imaging in deep tissue, provided that the generated photoacoustic signal is sufficiently strong.


Asunto(s)
Acústica , Fenómenos Ópticos , Espectrometría Raman , Aceite Mineral/química , Ultrasonido
16.
J Biophotonics ; 14(4): e202000384, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33438837

RESUMEN

Dysfunctional mitochondrial activity can lead to a variety of different diseases. As such, there exists a need to quantify changes in mitochondria function as it relates to these specific diseased states. Here, we present the use of resonance Raman (RR) spectroscopy as a tool to determine changes in isolated mitochondrial activity. RR spectroscopy, using 532 nm as the excitation source, specifically provides information on the reduction and oxidation (RedOx) state of cytochrome c, which is determined by the activity of protein complexes in the electron transport chain (ETC). In this model, injection of the substrate succinate into the mitochondrial sample is used to drive the ETC, which causes a subsequent change in cytochrome c RedOx state. This change in RedOx state is tracked by RR spectroscopy. This tool gives real-time information on the rise and fall of the amount of reduced cytochrome c within the mitochondrial sample, providing a method for rapid assessment of mitochondrial metabolism that has broad applications in both basic science and medical research.


Asunto(s)
Citocromos c , Mitocondrias , Animales , Citocromos c/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Espectrometría Raman , Porcinos
17.
PLoS Negl Trop Dis ; 15(4): e0009308, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33793555

RESUMEN

Mayaro virus (MAYV) is an alphavirus endemic to South and Central America associated with sporadic outbreaks in humans. MAYV infection causes severe joint and muscle pain that can persist for weeks to months. Currently, there are no approved vaccines or therapeutics to prevent MAYV infection or treat the debilitating musculoskeletal inflammatory disease. In the current study, a prophylactic MAYV vaccine expressing the complete viral structural polyprotein was developed based on a non-replicating human adenovirus V (AdV) platform. Vaccination with AdV-MAYV elicited potent neutralizing antibodies that protected WT mice against MAYV challenge by preventing viremia, reducing viral dissemination to tissues and mitigating viral disease. The vaccine also prevented viral-mediated demise in IFN⍺R1-/- mice. Passive transfer of immune serum from vaccinated animals similarly prevented infection and disease in WT mice as well as virus-induced demise of IFN⍺R1-/- mice, indicating that antiviral antibodies are protective. Immunization with AdV-MAYV also generated cross-neutralizing antibodies against two related arthritogenic alphaviruses-chikungunya and Una viruses. These cross-neutralizing antibodies were protective against lethal infection in IFN⍺R1-/- mice following challenge with these heterotypic alphaviruses. These results indicate AdV-MAYV elicits protective immune responses with substantial cross-reactivity and protective efficacy against other arthritogenic alphaviruses. Our findings also highlight the potential for development of a multi-virus targeting vaccine against alphaviruses with endemic and epidemic potential in the Americas.


Asunto(s)
Adenoviridae/genética , Alphavirus/inmunología , Fiebre Chikungunya/prevención & control , Virus Chikungunya/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Protección Cruzada/inmunología , Modelos Animales de Enfermedad , Femenino , Ingeniería Genética/métodos , Vectores Genéticos/genética , Inmunización , Masculino , Ratones , Ratones Endogámicos C57BL , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
18.
Biomed Opt Express ; 12(11): 7082-7091, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34858701

RESUMEN

The electronic or molecular mechanisms that initiate photobiomodulation (PBM) in cells are not yet fully understood. The porcine complex III (C-III) of the electron transport chain was characterized with transient absorption spectroscopy (TAS). We then applied our recently developed continuous wave laser coupled TAS procedure (CW-TAS) to investigate the effect of red light irradiances on the heme dynamics of C-III in its c1 reduced state. The time constants were found to be 3.3 ± 0.3 ps for vibrational cooling of the oxidized state and 4.9 ± 0.4 ps for rebinding of the photodissociated axial ligand of the c1 reduced state. The analysis of the CW-TAS procedure yielded no significant changes in the C-III heme dynamics. We rule out the possibility of 635 nm CW light at 4.7 mW/cm2 inducing a PBM effect on the heme dynamic of C-III, specifically with the photodissociation of its axial ligand.

19.
J Photochem Photobiol B ; 222: 112271, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34364080

RESUMEN

Photobiomodulation (PBM) describes the use of low irradiance light in the red to near-infrared wavelength range to stimulate biological effects in tissue, and many biological and spectroscopic techniques are used to study PBM. However, these techniques focus on the products or downstream effects rather than the electronic transitions that initiate the PBM processes. This study presents a novel approach to studying low irradiance light exposures on individual proteins and/or protein complexes by combining a continuous wave (CW) laser diode with femtosecond transient absorption spectroscopy (TAS), coined here as CW-TAS, and tests the system on reduced cytochrome c (Cyt c) for proof of principle. TAS was conducted using a 532-nm excitation pump beam and a 350-600 nm supercontinuum probe. CW laser diodes with wavelengths of 450 nm, 635 nm, and 808 nm were interchangeably fiber coupled into the HELIOS Fire. Samples of Cyt c were tested by TAS using a pump power of 15 µW, both with and without CW exposure. CW exposures were carried out with irradiances of 1.60 and 3.20 mW/cm2, except for 808 nm, which was only tested at 1.60 mW/cm2. Both kinetic and global analyses were performed on the TAS data and the time constants for sets with and without CW exposures were compared. The TAS data for Cyt c with the full dosage of CW exposures did not alter the TAS data distinguishably from the control data. No new electronic transient signals were observed beyond the background when testing Cyt c with the CW exposures. Kinetic analysis confirmed that existing transients did not deviate beyond uncertainty. Global time constants for Cyt c were calculated to be 0.25 ± 0.03 ps and 5.1 ± 0.3 ps for the control study, and the time constants for the CW exposed Cyt c were not significantly different. This study concludes that CW irradiation, at doses delivered, does not alter the transient absorption data of Cyt c. The CW-TAS method provides a new tool for studying PBM effects in other proteins and protein complexes that might respond to the CW wavelengths, such as Complex IV, in future studies.


Asunto(s)
Láseres de Semiconductores , Espectrofotometría/métodos , Citocromos c/química , Cinética , Luz , Oxidación-Reducción
20.
Virology ; 557: 23-33, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33601113

RESUMEN

Herpesviruses encode multiple glycoproteins required for different stages of viral attachment, fusion, and envelopment. The protein encoded by the human cytomegalovirus (HCMV) open reading frame UL116 forms a stable complex with glycoprotein H that is incorporated into virions. However, the function of this complex remains unknown. Herein, we characterize R116, the rat CMV (RCMV) putative homolog of UL116. Two R116 transcripts were identified in fibroblasts with three proteins expressed with molecular weights of 42, 58, and 82 kDa. R116 is N-glycosylated, expressed with late viral gene kinetics, and is incorporated into the virion envelope. RCMV lacking R116 failed to result in productive infection of fibroblasts and siRNA knockdown of R116 substantially reduced RCMV infectivity. Complementation in trans of an R116-deficient virus restored ability of the virus to infect fibroblasts. Finally, UL116 knockdown also decreased HCMV infectivity indicating that R116 and UL116 both contribute to viral infectivity.


Asunto(s)
Citomegalovirus/genética , Fibroblastos/virología , Sistemas de Lectura Abierta/genética , Proteínas del Envoltorio Viral/genética , Virión/química , Animales , Citomegalovirus/química , Glicosilación , Humanos , ARN Bicatenario , Ratas , Acoplamiento Viral , Internalización del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA