Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Int J Mol Sci ; 24(20)2023 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-37895108

RESUMO

Photobiomodulation (PBM) is the regulation of biological processes using light energy from sources such as lasers or light-emitting diodes. Components of the nervous system, such as the brain and peripheral nerves, are important candidate PBM targets due to the lack of therapeutic modalities for the complete cure of neurological diseases. PBM can be applied either to regenerate damaged organs or to prevent or reduce damage caused by disease. Although recent findings have suggested that neural cells can be regenerated, which contradicts our previous understanding, neural structures are still thought to have weaker regenerative capacity than other systems. Therefore, enhancing the regenerative capacity of the nervous system would aid the future development of therapeutics for neural degeneration. PBM has been shown to enhance cell differentiation from stem or progenitor cells to near-target or target cells. In this review, we have reviewed research on the effects of PBM on neurogenesis in the central nervous system (e.g., animal brains) and the peripheral nervous system (e.g., peripheral sensory neural structures) and sought its potential as a therapeutic tool for intractable neural degenerative disorders.


Assuntos
Terapia com Luz de Baixa Intensidade , Células-Tronco , Animais , Neurogênese , Encéfalo , Diferenciação Celular , Sistema Nervoso Periférico
2.
Biochem Biophys Res Commun ; 545: 33-39, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33535104

RESUMO

Zika virus (ZIKV) infection in both infants and adults is associated with neurological complications including, but not limited to, microcephaly and Guillain-Barre syndrome. Antibody therapy can be effective against virus infection. We isolated ZIKV envelope domain III-specific neutralizing antibodies (nAbs) from two convalescent patients with ZIKV infection. One antibody, 2F-8, exhibited potent in vitro neutralizing activity against Asian and American strains of ZIKV. To prevent FcγR-mediated antibody-dependent enhancement, we prepared IgG1 with LALA variation. A single dose of 2F-8 in the context of IgG1 or IgG1-LALA prior to or post lethal ZIKV challenge conferred complete protection in mice.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Proteínas do Envelope Viral/imunologia , Zika virus/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Especificidade de Anticorpos , Modelos Animais de Doenças , Feminino , Humanos , Técnicas In Vitro , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Testes de Neutralização , Gravidez , Domínios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas do Envelope Viral/química , Zika virus/química , Infecção por Zika virus/imunologia , Infecção por Zika virus/prevenção & controle
3.
Lasers Med Sci ; 36(9): 1917-1925, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33604771

RESUMO

Photobiomodulation (PBM) is a therapeutic approach to certain diseases based on light energy. Currently, stem cells (SCs) are being considered as putative treatments for previously untreatable diseases. One medical condition that could be treated using SCs is sensorineural hearing loss. Theoretically, if properly delivered and differentiated, SCs could replace lost hair cells in the cochlea. However, this is not currently possible due to the structural complexity and limited survival of SCs within the cochlea. PBM facilitates SC differentiation into other target cells in multiple lineages. Using light with a wavelength > 800 nm, which can penetrate the inner ear through the tympanic membrane, we assessed morphological changes of mouse embryonic stem cells (mESCs) during "otic organoid" generation, and within the scala media (SM) of the cochlea, after light energy stimulation. We observed enhanced differentiation, which was confirmed by an increased number of otic vesicles and increased cell attachment inside the SM. These results suggest that > 800-nm light affected the morphology of mESCs within otic organoids and SM of the cochlea. Based on our results, light energy could be used to enhance otic sensory differentiation, despite the structural complexity of the inner ear and limited survival time of SCs within the cochleae. Additional studies to refine the light energy delivery technology and maximize the effect on otic differentiation are required.


Assuntos
Orelha Interna , Organoides , Animais , Diferenciação Celular , Cóclea , Ducto Coclear , Camundongos , Células-Tronco
4.
Lasers Med Sci ; 36(9): 1941-1947, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33822307

RESUMO

Sensorineural hearing loss is an intractable disease. Acoustic overstimulation creates hearing loss; many patients exhibit social and emotional dysfunctions. In a model of noise-induced hearing loss (NIHL), low-level laser photobiomodulation (PBM) at a near-infrared wavelength significantly improved auditory brainstem response (ABR) thresholds. In addition, both N-acetyl-L-cysteine (NAC) and acetyl-L-carnitine (ALCAR) attenuated NIHL, reducing the effects of noise trauma in the cochlea and the central auditory system. Here, we combined PBM with antioxidants to explore hearing threshold recovery and morphological hair cell changes after rats were exposed to noise. The average auditory brainstem response thresholds after PBM/NAC combination treatment were reduced from the apex to the basal turn at all of 8, 16, and 32 kHz compared to the noise-only group. The PBM/NAC combination treated group exhibited intact outer hair cells in all turns, and significantly greater hair cell numbers in the middle and basal cochlear turns, than did controls. Thus, PBM/NAC treatment may prevent hearing dysfunction caused by NIHL.


Assuntos
Perda Auditiva Provocada por Ruído , Acetilcisteína/farmacologia , Animais , Limiar Auditivo , Cóclea , Potenciais Evocados Auditivos do Tronco Encefálico , Células Ciliadas Auditivas , Perda Auditiva Provocada por Ruído/tratamento farmacológico , Humanos , Ratos
5.
Int J Mol Sci ; 22(19)2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34638648

RESUMO

Otic organoids have the potential to resolve current challenges in hearing loss research. The reproduction of the delicate and complex structure of the mammalian cochlea using organoids requires high efficiency and specificity. Recent attempts to strengthen otic organoids have focused on the effects of the Wnt signaling pathway on stem cell differentiation. One important aspect of this is the evaluation of undesirable effects of differentiation after Wnt activation. In the present study, we differentiated mouse embryonic stem cell embryoid bodies (EB) into otic organoids and observed two morphologies with different cell fates. EBs that underwent a core ejection process, or 'enucleation,' were similar to previously reported inner ear organoids. Meanwhile, EBs that retained their core demonstrated features characteristic of neural organoids. The application of a Wnt agonist during the maturation phase increased enucleation, as well as otic organoid formation, in turn leading to sensory hair cell-like cell generation. However, with a longer incubation period, Wnt activation also led to EBs with 'beating' organoids that exhibited spontaneous movement. This observation emphasizes the necessity of optimizing Wnt enhancement for the differentiation of specific cells, such as those found in the inner ear.


Assuntos
Diferenciação Celular/fisiologia , Cóclea/metabolismo , Cóclea/fisiologia , Organoides/metabolismo , Organoides/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Células Cultivadas , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/fisiologia , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/fisiologia , Células-Tronco Pluripotentes/metabolismo
7.
Audiol Neurootol ; 24(2): 100-108, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31207595

RESUMO

BACKGROUND: Endoscopic ear surgery has recently increased, but it is still inconvenient and time-consuming to place packing material in the middle ear with one hand. Poloxamer 407 (P407) is a thermo-reversible gel that can be easily administered with one hand into the middle ear cavity in liquid form. Upon warming to body temperature, the gel form of P407 can support the graft in the target position and is known to prevent postsurgical tissue adhesion. OBJECTIVES: We aim to investigate the feasibility of P407 as packing material in an animal model. Male Hartley guinea pigs (350 and 400 g) were utilized in this study. METHOD: The animals were randomly divided into 3 groups according to the packing material: the control group, the P407 group, and the gelatin group. To assess the role of packing material on bacterial colonization, left ears were inoculated with Streptococcus pneumoniae through the tympanic membrane using a 0° endoscope. Five days after inoculation, the middle ear cavity was packed through a transbullar approach using 18% P407 or gelatin in both ears. In the control group, no ear pack was inserted. The tympanic membrane was examined every week using a 0° 1.9-mm endoscope until 6 weeks. Half of the animals in each group were sacrificed 6 weeks after placement of the packing materials. RESULTS: Compared with the absorbable gelatin sponge, the P407 group showed little inflammation or fibrosis in the tympanic membrane and middle ear mucosa regardless of bacterial inoculation. The gelatin group showed severe otorrhea or perforation until 2 weeks in the right ear (2 of 4) and the left ear (1 of 4). Even though the endoscopic findings were similar between both packing groups at 6 weeks, histological analysis showed persistent packing material, inflammatory cells, and fibrosis in the gelatin group compared to the P407 group. CONCLUSIONS: This study suggested that P407 is feasible as a packing material to handle with one hand and to prevent adhesion, especially in infected middle ear mucosa. Although there is a lack of data on how well P407 supports grafts, we suggest that P407 could be a candidate for packing material in endoscopic ear surgery.


Assuntos
Orelha Média/cirurgia , Otoscopia , Poloxâmero , Animais , Modelos Animais de Doenças , Gelatina , Esponja de Gelatina Absorvível , Cobaias , Masculino , Aderências Teciduais/prevenção & controle , Membrana Timpânica/patologia
8.
Lasers Med Sci ; 34(2): 367-375, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30105484

RESUMO

Gene therapy is the delivery of a therapeutic gene into target cells to treat disorders by replacing disease-causing mutated genes with healthy ones. Gene therapy of the inner ear has been recently described, with applications for sensorineural hearing loss. However, gene delivery to the location of the inner ear, and thus efficacy of therapy, is challenging. Photobiomodulation (PBM) with a low-level laser has been suggested to have a therapeutic effect and has the potential to augment gene therapy. To investigate whether PBM improves the rate of adenovirus (Ad)-mediated viral delivery, we compared low-level laser therapy (LLLT) and non-LLLT HEI-OC1 cells treated with an Ad viral vector carrying green fluorescent protein (GFP). Cultured HEI-OC1 cells were divided into six groups: no treatment control, LLLT only, 1 µL Ad-GFP, 3 µL Ad-GFP, 1 µL Ad-GFP + LLLT, and 3 µL Ad-GFP + LLLT (LLLT: 808 nm at 15 mW for 15 min). Cells were irradiated twice: at 2 h and again at 24 h. A nonparametric Mann-Whitney U test was used to statistically analyze differences between the control and treatment groups. The viral inoculations used in this study did not change the amount of viable HEI-OC1 cells (N = 4-8). The 1 µL Ad-GFP + LLLT and 3 µL Ad-GFP + LLLT groups showed an increased density of GFP-positive cells compared to 1 µL and 3 µL Ad-GFP cells (N = 5-8, 1 µL: p = 0.0159; 3 µL: p = 0.0168,). The quantitative analysis of the epifluorescence of the 1 µL Ad-GFP + LLLT, and 3 µL Ad-GFP + LLLT groups revealed increased GFP expression/cell compared to 1 µL and 3 µL Ad-GFP cells (N = 6-15, 1 µL: p = 0.0082; 3 µL: p = 0.0012). The RT-qPCR results were consistent (N = 4-5, p = 0.0159). These findings suggest that PBM may enhance the gene delivery of Ad-mediated viral transduction, and the combination of the two may be a promising tool for gene therapy for sensorineural hearing loss.


Assuntos
Adenoviridae/metabolismo , Células Ciliadas Auditivas/metabolismo , Terapia com Luz de Baixa Intensidade , Transdução Genética/métodos , Animais , Linhagem Celular , Sobrevivência Celular , Fluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos
9.
Int J Mol Sci ; 20(20)2019 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-31614869

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) induces severe aggravating respiratory failure in infected patients, frequently resulting in mechanical ventilation. As limited therapeutic antibody is accumulated in lung tissue following systemic administration, inhalation is newly recognized as an alternative, possibly better, route of therapeutic antibody for pulmonary diseases. The nebulization process, however, generates diverse physiological stresses, and thus, the therapeutic antibody must be resistant to these stresses, remain stable, and form minimal aggregates. We first isolated a MERS-CoV neutralizing antibody that is reactive to the receptor-binding domain (RBD) of spike (S) glycoprotein. To increase stability, we introduced mutations into the complementarity-determining regions (CDRs) of the antibody. In the HCDRs (excluding HCDR3) in this clone, two hydrophobic residues were replaced with Glu, two residues were replaced with Asp, and four residues were replaced with positively charged amino acids. In LCDRs, only two Leu residues were replaced with Val. These modifications successfully generated a clone with significantly greater stability and equivalent reactivity and neutralizing activity following nebulization compared to the original clone. In summary, we generated a MERS-CoV neutralizing human antibody that is reactive to recombinant MERS-CoV S RBD protein for delivery via a pulmonary route by introducing stabilizing mutations into five CDRs.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Regiões Determinantes de Complementaridade/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Administração por Inalação , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/genética , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/genética , Chlorocebus aethiops , Regiões Determinantes de Complementaridade/genética , Humanos , Células Vero
10.
Clin Infect Dis ; 63(3): 363-9, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27090992

RESUMO

BACKGROUND: The largest outbreak of Middle East respiratory syndrome coronavirus (MERS-CoV) outside the Middle East occurred in South Korea in 2015 and resulted in 186 laboratory-confirmed infections, including 36 (19%) deaths. Some hospitals were considered epicenters of infection and voluntarily shut down most of their operations after nearly half of all transmissions occurred in hospital settings. However, the ways that MERS-CoV is transmitted in healthcare settings are not well defined. METHODS: We explored the possible contribution of contaminated hospital air and surfaces to MERS transmission by collecting air and swabbing environmental surfaces in 2 hospitals treating MERS-CoV patients. The samples were tested by viral culture with reverse transcription polymerase chain reaction (RT-PCR) and immunofluorescence assay (IFA) using MERS-CoV Spike antibody, and electron microscopy (EM). RESULTS: The presence of MERS-CoV was confirmed by RT-PCR of viral cultures of 4 of 7 air samples from 2 patients' rooms, 1 patient's restroom, and 1 common corridor. In addition, MERS-CoV was detected in 15 of 68 surface swabs by viral cultures. IFA on the cultures of the air and swab samples revealed the presence of MERS-CoV. EM images also revealed intact particles of MERS-CoV in viral cultures of the air and swab samples. CONCLUSIONS: These data provide experimental evidence for extensive viable MERS-CoV contamination of the air and surrounding materials in MERS outbreak units. Thus, our findings call for epidemiologic investigation of the possible scenarios for contact and airborne transmission, and raise concern regarding the adequacy of current infection control procedures.


Assuntos
Infecções por Coronavirus/transmissão , Surtos de Doenças , Microbiologia Ambiental , Coronavírus da Síndrome Respiratória do Oriente Médio/isolamento & purificação , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/virologia , Hospitais , Humanos , Controle de Infecções , República da Coreia/epidemiologia
11.
Biochem Biophys Res Commun ; 478(4): 1594-601, 2016 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-27586275

RESUMO

Various influenza virus entry inhibitors are being developed as therapeutic antiviral agents in ongoing preparation for emerging influenza viruses, particularly those that may possess drug resistance to the current FDA-approved neuraminidase inhibitors. In this study, small molecules having the pyrrolopyridinamine (PPA), aminothiadiazole (ATD), dihydrofuropyridine carboxamide (HPC), or imidazopyridinamine (IPA) moiety were selected from a target-focused chemical library for their inhibitory activity against influenza A virus by high-throughput screening using the PR8GFP assay. Activity was evaluated by measuring changes the proportion of GFP-expressing cells as a reflection of influenza virus infection. Among them, PPA showed broad-spectrum activity against multiple influenza A viruses and influenza B virus. PPA was found to block the early stages of influenza virus infection using a time-of-addition assay. Using additional phenotypic assays that dissect the virus entry process, it appears that the antiviral activity of PPA against influenza virus can be attributed to interference of the post-fusion process: namely, virus uncoating and nuclear import of viral nucleoprotein complexes. Based on these results, PPA is an attractive chemical moiety that can be used to develop new antiviral drug candidates against influenza viruses.


Assuntos
Antivirais/farmacologia , Orthomyxoviridae/efeitos dos fármacos , Piridinas/farmacologia , Pirróis/farmacologia , Internalização do Vírus/efeitos dos fármacos , Células A549 , Animais , Aves/virologia , Morte Celular/efeitos dos fármacos , Cães , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Influenza Aviária/virologia , Células Madin Darby de Rim Canino , Proteínas Nucleares/metabolismo , Piridinas/química , Pirróis/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
12.
Biochem Biophys Res Commun ; 471(2): 282-9, 2016 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-26850850

RESUMO

The need to develop anti-influenza drugs with novel antiviral mechanisms is urgent because of the rapid rate of antigenic mutation and the emergence of drug-resistant viruses. We identified a novel anti-influenza molecule by screening 861 plant-derived natural components using a high-throughput image-based assay that measures inhibition of the influenza virus infection. 1,3,4,6-tetra-O-galloyl-ß-D-glucopyranoside (TGBG) from Euphorbia humifusa Willd showed broad-spectrum anti-influenza activity against two seasonal influenza A strains, A/California/07/2009 (H1N1) and A/Perth/16/2009 (H3N2), and seasonal influenza B strain B/Florida/04/2006. We investigated the mode of action of TGBG using neuraminidase activity inhibition and time-of-addition assays, which evaluate the viral release and entry steps, respectively. We found that TGBG exhibits a novel antiviral mechanism that differs from the FDA-approved anti-influenza drugs oseltamivir which inhibits viral release, and amantadine which inhibits viral entry. Immunofluorescence assay demonstrated that TGBG significantly inhibits nuclear export of influenza nucleoproteins (NP) during the early stages of infection causing NP to accumulate in the nucleus. In addition, influenza-induced activation of the Akt signaling pathway was suppressed by TGBG in a dose-dependent manner. These data suggest that a putative mode of action of TGBG involves inhibition of viral ribonucleoprotein (vRNP) export from the nucleus to the cytoplasm consequently disrupting the assembly of progeny virions. In summary, TGBG has potential as novel anti-influenza therapeutic with a novel mechanism of action.


Assuntos
Antivirais/administração & dosagem , Núcleo Celular/metabolismo , Euphorbia/química , Vírus da Influenza A/fisiologia , Vírus da Influenza B/fisiologia , Ribonucleoproteínas/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Núcleo Celular/efeitos dos fármacos , Cães , Relação Dose-Resposta a Droga , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Extratos Vegetais/administração & dosagem , Internalização do Vírus/efeitos dos fármacos
13.
Lasers Med Sci ; 31(2): 323-33, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26738500

RESUMO

We evaluated functional and morphological changes after trans-tympanic laser application using several different powers of photobiomodulation (PBM). The left (L) ears of 17 rats were irradiated for 30 min daily over 14 days using a power density of 909.1 (group A, 5040 J), 1136.4 (group B, 6300 J), and 1363.6 (group C, 7560 J) mW/cm(2). The right (N) ears served as controls. The safety of PBM was determined by endoscopic findings, auditory brainstem response (ABR) thresholds, and histological images of hair cells using confocal microscopy, and light microscopic images of the external auditory canal (EAC) and tympanic membrane (TM). Endoscopic findings revealed severe inflammation in the TM of C group; no other group showed damage in the TM. No significant difference in ABR threshold was found in the PBM-treated groups (excluding the group with TM damage). Confocal microscopy showed no histological difference between the AL and AN, or BL and BN groups. However, light microscopy showed more prominent edema, inflammation, and vascular congestion in the TM of BL ears. This study found a dose-response relationship between laser power parameters and TM changes. These results will be useful for defining future allowance criteria for trans-tympanic laser therapies.


Assuntos
Terapia com Luz de Baixa Intensidade/efeitos adversos , Segurança , Membrana Timpânica/efeitos da radiação , Animais , Meato Acústico Externo/fisiologia , Meato Acústico Externo/efeitos da radiação , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos da radiação , Masculino , Ratos , Membrana Timpânica/fisiologia
14.
Nat Commun ; 15(1): 2619, 2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38521764

RESUMO

Immunity to infectious diseases is predominantly studied by measuring immune responses towards a single pathogen, although co-infections are common. In-depth mechanisms on how co-infections impact anti-viral immunity are lacking, but are highly relevant to treatment and prevention. We established a mouse model of co-infection with unrelated viruses, influenza A (IAV) and Semliki Forest virus (SFV), causing disease in different organ systems. SFV infection eight days before IAV infection results in prolonged IAV replication, elevated cytokine/chemokine levels and exacerbated lung pathology. This is associated with impaired lung IAV-specific CD8+ T cell responses, stemming from suboptimal CD8+ T cell activation and proliferation in draining lymph nodes, and dendritic cell paralysis. Prior SFV infection leads to increased blood brain barrier permeability and presence of IAV RNA in brain, associated with increased trafficking of IAV-specific CD8+ T cells and establishment of long-term tissue-resident memory. Relative to lung IAV-specific CD8+ T cells, brain memory IAV-specific CD8+ T cells have increased TCR repertoire diversity within immunodominant DbNP366+CD8+ and DbPA224+CD8+ responses, featuring suboptimal TCR clonotypes. Overall, our study demonstrates that infection with an unrelated neurotropic virus perturbs IAV-specific immune responses and exacerbates IAV disease. Our work provides key insights into therapy and vaccine regimens directed against unrelated pathogens.


Assuntos
Coinfecção , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Vírus , Camundongos , Animais , Humanos , Influenza Humana/patologia , Linfócitos T CD8-Positivos , Coinfecção/patologia , Receptores de Antígenos de Linfócitos T , Pulmão/patologia
15.
Sci Rep ; 13(1): 6220, 2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37069238

RESUMO

Photobiomodulation (PBM) is a therapeutic tool that uses red or near-infrared light in medical applications. It's applications in both central (CNS) and peripheral nervous system (PNS) are widely studied. Among glial cells, astrocytes are known to be activated in injured or damaged brains. Astrocytic cell migration is crucial for maintaining homeostasis in the brain. Our previous study showed that PBM led to astrocyte proliferation and differentiation, but the effects on migration has not been investigated. The aim of this study was to evaluate the effect of PBM on astrocyte migration, drebrin (DBN) expression and cytoplasmic morphology using primary cultured rat astrocyte. We applied a 660-nm light-emitting diode (LED) with fluence of 6, 12 and 18 J/cm2. PBM effects on astrocyte migration were analyzed by two different migration assays (scratch assay and transwell assay). We used immunofluorescence microscopy for visualizing DBN and glial-fibrillary acidic protein (GFAP) and analysis of DBN expression and astrocyte cytoplasmic morphology. Both scratch assay and transwell assay showed significant difference in astrocyte migration following PBM irradiation. With these specific fluence conditions, differences in DBN expression and cell morphology were revealed. PBM could increase the astrocyte migration by altering the cell morphology and DBN expression pattern.


Assuntos
Astrócitos , Encéfalo , Ratos , Animais , Astrócitos/metabolismo , Proliferação de Células , Movimento Celular
16.
Cells ; 12(8)2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37190055

RESUMO

Diabetes mellitus contributes to 15-25% of all chronic foot ulcers. Peripheral vascular disease is a cause of ischemic ulcers and exacerbates diabetic foot disease. Cell-based therapies are viable options to restore damaged vessels and induce the formation of new vessels. Adipose-derived stem cells (ADSCs) have the potential for angiogenesis and regeneration because of their greater paracrine effect. Preclinical studies are currently using other forced enhancement techniques (e.g., genetic modification or biomaterials) to increase the efficacy of human ADSC (hADSC) autotransplantation. Unlike genetic modifications and biomaterials, many growth factors have been approved by the equivalent regulatory authorities. This study confirmed the effect of enhanced human ADSC (ehADSC)s with a cocktail of FGF and other pharmacological agents to promote wound healing in diabetic foot disease. In vitro, ehADSCs exhibited a long and slender spindle-shaped morphology and showed significantly increased proliferation. In addition, it was shown that ehADSCs have more functionalities in oxidative stress toleration, stem cell stemness, and mobility. In vivo, the local transplantation of 1.2 × 106 hADSCs or ehADSCs was performed in animals with diabetes induced by STZ. The ehADSC group showed a statistically decreased wound size and increased blood flow compared with the hADSC group and the sham group. Human Nucleus Antigen (HNA) positive cells were observed in some ADSC-transplanted animals. The ehADSC group showed a relatively higher portion of HNA-positive animals than the hADSC group. The blood glucose levels showed no significant difference among the groups. In conclusion, the ehADSCs showed a better performance in vitro, compared with conventional hADSCs. Additionally, a topical injection of ehADSCs into diabetic wounds enhanced wound healing and blood flow, while improving histological markers suggesting revascularization.


Assuntos
Diabetes Mellitus Experimental , Pé Diabético , Humanos , Ratos , Animais , Estreptozocina , Tecido Adiposo , Pé Diabético/terapia , Pé Diabético/patologia , Fatores de Crescimento de Fibroblastos/farmacologia , Cicatrização/fisiologia , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/patologia , Células-Tronco , Materiais Biocompatíveis/farmacologia
17.
Stem Cells Int ; 2023: 6845571, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38020205

RESUMO

Sensorineural hearing loss is very difficult to treat. Currently, one of the techniques used for hearing rehabilitation is a cochlear implant that can transform sound into electrical signals instead of inner ear hair cells. However, the prognosis remains very poor if sufficient auditory nerve cells are not secured. In this study, the effect of mouse embryonic stem cells (mESC) and photobiomodulation (PBM) combined treatment on auditory function and auditory nerve cells in a secondary neuropathy animal model was investigated. To confirm the engraftment of stem cells in vitro, cochlear explants were treated with kanamycin (KM) to mimic nerve damage and then cocultured with GFP-mESC. GFP-mESCs were observed to have attached and integrated into the explanted samples. An animal model for secondary neurodegeneration was achieved by KM treatment and was treated by a combination therapy of GFP-mESC and NIR-PBM at 8 weeks of KM treatment. Hearing recovery by functional testing using auditory brain stem response (ABR) and eABR was measured as well as morphological changes and epifluorescence analysis were conducted after 2 weeks of combination therapy. KM treatment elevated the hearing threshold at 70-80 dB and even after the combination treatment with GFP-mESC and PBM was applied, the auditory function was not restored. In addition, the stem cells transplanted into cochlea has exponentially increased due to PBM treatment although did not produce any malignancy. This study confirmed that the combined treatment with mESC and PBM could not improve hearing or increase the response of the auditory nerve. Nevertheless, it is noteworthy in this study that the cells are distributed in most cochlear tissues and the proliferation of stem cells was very active in animals irradiated with PBM compared to other groups wherein the stem cells had disappeared immediately after transplantation or existed for only a short period of time.

18.
PLoS One ; 18(5): e0284626, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37216352

RESUMO

Noise exposure can destroy the synaptic connections between hair cells and auditory nerve fibers without damaging the hair cells, and this synaptic loss could contribute to difficult hearing in noisy environments. In this study, we investigated whether delivering lithium chloride to the round-window can regenerate synaptic loss of cochlea after acoustic overexposure. Our rat animal model of noise-induced cochlear synaptopathy caused about 50% loss of synapses in the cochlear basal region without damaging hair cells. We locally delivered a single treatment of poloxamer 407 (vehicle) containing lithium chloride (either 1 mM or 2 mM) to the round-window niche 24 hours after noise exposure. Controls included animals exposed to noise who received only the vehicle. Auditory brainstem responses were measured 3 days, 1 week, and 2 weeks post-exposure treatment, and cochleas were harvested 1 week and 2 weeks post-exposure treatment for histological analysis. As documented by confocal microscopy of immunostained ribbon synapses, local delivery of 2 mM lithium chloride produced synaptic regeneration coupled with corresponding functional recovery, as seen in the suprathreshold amplitude of auditory brainstem response wave 1. Western blot analyses revealed that 2 mM lithium chloride suppressed N-methyl-D-aspartate (NMDA) receptor expression 7 days after noise-exposure. Thus, round-window delivery of lithium chloride using poloxamer 407 reduces cochlear synaptic loss after acoustic overexposure by inhibiting NMDA receptor activity in rat model.


Assuntos
Perda Auditiva Provocada por Ruído , Receptores de N-Metil-D-Aspartato , Ratos , Animais , Receptores de N-Metil-D-Aspartato/metabolismo , Cloreto de Lítio , Perda Auditiva Provocada por Ruído/etiologia , Poloxâmero , Limiar Auditivo/fisiologia , Cóclea/patologia , Sinapses/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia
19.
Cell Rep Med ; 4(4): 101017, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37030296

RESUMO

Immunocompromised hematology patients are vulnerable to severe COVID-19 and respond poorly to vaccination. Relative deficits in immunity are, however, unclear, especially after 3 vaccine doses. We evaluated immune responses in hematology patients across three COVID-19 vaccination doses. Seropositivity was low after a first dose of BNT162b2 and ChAdOx1 (∼26%), increased to 59%-75% after a second dose, and increased to 85% after a third dose. While prototypical antibody-secreting cells (ASCs) and T follicular helper (Tfh) cell responses were elicited in healthy participants, hematology patients showed prolonged ASCs and skewed Tfh2/17 responses. Importantly, vaccine-induced expansions of spike-specific and peptide-HLA tetramer-specific CD4+/CD8+ T cells, together with their T cell receptor (TCR) repertoires, were robust in hematology patients, irrespective of B cell numbers, and comparable to healthy participants. Vaccinated patients with breakthrough infections developed higher antibody responses, while T cell responses were comparable to healthy groups. COVID-19 vaccination induces robust T cell immunity in hematology patients of varying diseases and treatments irrespective of B cell numbers and antibody response.


Assuntos
COVID-19 , Neoplasias Hematológicas , Humanos , Receptores de Antígenos de Linfócitos T alfa-beta , Vacinas contra COVID-19 , SARS-CoV-2 , Vacina BNT162 , Linfócitos T CD8-Positivos
20.
Biomed Res Int ; 2021: 4956404, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34250085

RESUMO

Hearing loss is a sensory deprivation that can affect the quality of life. Currently, only rehabilitation devices such as hearing aids and cochlear implants are used, without a definitive cure. However, in chronic hearing-deprived patients, in whom secondary auditory neural degeneration is expected, a relatively poor rehabilitation prognosis is projected. Stem cell therapy for cochlear neural structures would be an easier and feasible strategy compared with cochlear sensory cells. Considering the highly developed cochlear implantation technology, improving cochlear neural health has significant medical and social effects. Stem cell delivery to Rosenthal's canal in an acutely damaged mouse model has been performed and showed cell survival and the possibility of differentiation. The results of stem cell transplantation in chronic auditory neural hearing loss should be evaluated because neural stem cell replacement therapy for chronic (long-term) sensorineural hearing loss is a major target in clinics. In the present study, we established a mouse model that mimicked chronic auditory neural hearing loss (secondary degeneration of auditory neurons after loss of sensory input). Then, mouse embryonic stem cells (mESCs) were transplanted into the scala tympani and survival and distribution of transplanted cells were compared between the acute and chronic auditory neural hearing loss models induced by ouabain or kanamycin (KM), respectively. The mESC survival was similar to the acute model, and perilymphatic distribution of cell aggregates was more predominant in the chronic model. Lastly, the effects of mESC transplantation on neural signal transduction observed in the cochlear nucleus (CN) were compared and a statistical increase was observed in the chronic model compared with other models. These results indicated that after transplantation, mESCs survived in the cochlea and increased the neural signaling toward the central auditory pathway, even in the chronic (secondary) hearing loss mouse model.


Assuntos
Vias Aferentes/patologia , Cóclea/patologia , Perda Auditiva Neurossensorial/terapia , Células-Tronco Embrionárias Murinas/transplante , Neurônios/patologia , Doença Aguda , Animais , Limiar Auditivo/fisiologia , Doença Crônica , Cóclea/fisiopatologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Perda Auditiva Neurossensorial/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA