Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Stem Cells ; 41(4): 307-309, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37098181
2.
Exp Eye Res ; 226: 109303, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36343671

RESUMEN

PURPOSE: Fuchs endothelial corneal dystrophy (FECD) is a progressive corneal disease that impacts the structure and stiffness of the Descemet's membrane (DM), the substratum for corneal endothelial cells (CECs). These structural alterations of the DM could contribute to the loss of the CECs resulting in corneal edema and blindness. Oxidative stress and transforming growth factor-ß (TGF-ß) pathways have been implicated in endothelial cell loss and endothelial to mesenchymal transition of CECs in FECD. Ascorbic acid (AA) is found at high concentrations in FECD and its impact on CEC survival has been investigated. However, how TGF-ß and AA effect the composition and rigidity of the CEC's matrix remains unknown. METHODS: In this study, we investigated the effect of AA, TGF-ß1 and TGF-ß3 on the deposition, ultrastructure, stiffness, and composition of the extracellular matrix (ECM) secreted by primary bovine corneal endothelial cells (BCECs). RESULTS: Immunofluorescence and electron microscopy post-decellularization demonstrated a robust deposition and distinct structure of ECM in response to treatments. AFM measurements showed that the modulus of the matrix in BCECs treated with TGF-ß1 and TGF-ß3 was significantly lower than the controls. There was no difference in the stiffness of the matrix between the AA-treated cell and controls. Gene Ontology analysis of the proteomics results revealed that AA modulates the oxidative stress pathway in the matrix while TGF-ß induces the expression of matrix proteins collagen IV, laminin, and lysyl oxidase homolog 1. CONCLUSIONS: Molecular pathways identified in this study demonstrate the differential role of soluble factors in the pathogenesis of FECD.


Asunto(s)
Distrofia Endotelial de Fuchs , Factor de Crecimiento Transformador beta1 , Animales , Bovinos , Factor de Crecimiento Transformador beta1/metabolismo , Células Endoteliales/metabolismo , Factor de Crecimiento Transformador beta3/metabolismo , Distrofia Endotelial de Fuchs/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Endotelio Corneal/metabolismo
3.
Diagnostics (Basel) ; 12(4)2022 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-35453845

RESUMEN

Background: Novel, non-invasive diagnostic biomarkers that facilitate early intervention in head and neck cancer are urgently needed. Polyamine metabolites have been observed to be elevated in numerous cancer types and correlated with poor prognosis. The aim of this study was to assess the concentration of polyamines in the saliva and urine from head and neck cancer (HNC) patients, compared to healthy controls. Methods: Targeted metabolomic analysis was performed on saliva and urine from 39 HNC patient samples and compared to 89 healthy controls using a quantitative, targeted liquid chromatography mass spectrometry approach. Results: The metabolites N1-acetylspermine (ASP), N8-acetylspermidine (ASD) and N1,N12-diacetylspermine (DAS) were detected at significantly different concentrations in the urine of HNC patients as compared to healthy controls. Only ASP was detected at elevated levels in HNC saliva as compared to healthy controls. Conclusion: These data suggest that assessment of polyamine-based metabolite biomarkers within the saliva and urine warrants further investigation as a potential diagnostic in HNC patients.

4.
Laryngoscope ; 132(3): 523-527, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33988246

RESUMEN

OBJECTIVES/HYPOTHESIS: To evaluate the safety and potential efficacy of autologous muscle-derived cells (AMDCs) for the treatment of swallowing impairment following treatment for oropharynx cancer. STUDY DESIGN: Prospective, phase I, open label, clinical trial. METHODS: Oropharynx cancer survivors disease free ≥2 years post chemoradiation were recruited. All patients had swallowing impairment but were not feeding tube dependent (Functional Oral Intake Scale [FOIS] ≥ 5). Muscle tissue (50-250 mg) was harvested from the vastus lateralis and 150 × 106 AMDCs were prepared (Cook MyoSite Inc., Pittsburgh, PA). The cells were injected into four sites throughout the intrinsic tongue musculature. Participants were followed for 24 months. The primary outcome measure was safety. Secondary endpoints included objective measures on swallowing fluoroscopy, oral and pharyngeal pressure, and changes in patient-reported outcomes. RESULTS: Ten individuals were enrolled. 100% (10/10) were male. The mean age of the cohort was 65 (±8.87) years. No serious adverse event occurred. Mean tongue pressure increased significantly from 26.3 (±11.1) to 31.8 (±9.5) kPa (P = .017). The mean penetration-aspiration scale did not significantly change from 5.6 (±2.1) to 6.8 (±1.8), and the mean FOIS did not significantly change from 5.4 (±0.5) to 4.6 (±0.7). The incidence of pneumonia was 30% (3/10) and only 10% (1/10) experienced deterioration in swallowing function throughout 2 years of follow-up. The mean eating assessment tool (EAT-10) did not significantly change from 24.1 (±5.57) to 21.3 (±6.3) (P = .12). CONCLUSION: Results of this phase I clinical trial demonstrate that injection of 150 × 106 AMDCs into the tongue is safe and may improve tongue strength, which is durable at 2 years. A blinded placebo-controlled trial is warranted. LEVEL OF EVIDENCE: 3 Laryngoscope, 132:523-527, 2022.


Asunto(s)
Trasplante de Células/métodos , Trastornos de Deglución/terapia , Neoplasias de Cabeza y Cuello/complicaciones , Células Musculares/trasplante , Anciano , Trastornos de Deglución/etiología , Fluoroscopía/métodos , Humanos , Masculino , Manometría , Estudios Prospectivos
5.
Ann Transl Med ; 9(15): 1275, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34532412

RESUMEN

BACKGROUND: To evaluate whether subretinal or intravitreal injection of human CD34+ bone marrow-derived stem cells (BMSC) can have protective effects on retinal degeneration that may be enhanced by coadministration of exosomes harvested from human bone marrow mesenchymal stem cells (MSCs). METHODS: Human CD34+ cells were harvested from the mononuclear cell fraction of bone marrow using magnetic beads and labeled with EGFP. Exosomes were harvested from cultured human MSCs under hypoxic conditions. Royal College of Surgeons (RCS) 3-weeks-old rats, immunosuppressed with cyclosporine A, received subretinal or intravitreal injection of CD34+ cells (50,000 cells), CD34+ cells with exosomes (50,000 cells+10 µg), exosomes alone (10 µg), or PBS. Retinal function was examined using electroretinography (ERG), and the eyes were harvested for histologic and immunohistochemical analysis. RESULTS: The b-wave amplitude of ERG at 2 weeks after injection was significantly higher in eyes with subretinal or intravitreal CD34+ BMSC alone or in combination with exosomes when compared to PBS injected eyes or untreated contralateral eyes. At 4 weeks after injection, the ERG signal decreased in all groups but eyes with subretinal CD34+ BMSCs alone or combined with exosomes showed partially preserved ERG signal and preservation of the outer nuclear layer of the retina near the injection site on histology when compared to eyes with PBS injection. Immunohistochemical analysis identified the human cells in the outer retina. Subretinal or intravitreal exosome injection had no effect on retinal degeneration when administered alone or in combination with CD34+ cells. CONCLUSIONS: Both subretinal and intravitreal injection of human CD34+ BMSCs can provide functional rescue of degenerating retina, although the effects were attenuated over time in this rat model. Regional preservation of the outer retina can occur near the subretinal injection site of CD34+ cells. These results suggest that CD34+ cells may have therapeutic potential in retinal degeneration.

6.
Laryngoscope ; 131(6): 1223-1228, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32770795

RESUMEN

OBJECTIVE: Liquid thickeners are commonly recommended in individuals with dysphagia and recurrent aspiration as a strategy for pneumonia prevention. The goal of this study was to examine the effects of small amounts of aspirated liquid thickener on the lungs. STUDY DESIGN: Animal model. Prospective small animal clinical trial. METHODS: Adult Sprague Dawley rats (n = 19) were divided into two groups and underwent three intratracheal instillations of either xanthan gum-based nectar-thick water (0.1-0.25 mL/kg) or water-only control over the course of 8 days. Blood was collected from a peripheral vein on days 1 and 8 and submitted for complete blood count (CBC) analysis. Rats were euthanized 10 days after the last instillation, and the lungs were harvested. Histopathology was conducted on lung specimens by a blinded licensed veterinary pathologist and scored for evidence of lung injury and pneumonia. RESULTS: Fifteen animals (8 nectar-thickener group, 7 control group) survived until the endpoint of the study (day 18). Serum CBC did not show abnormalities at any timepoint in either group. Histological evidence of lung inflammation and edema were significantly greater in the nectar-thick group compared to controls (P < .05). Signs of inflammation included aggregates of foamy macrophages, expansion of bronchiolar lymphoid tissue, and large numbers of eosinophilic intraalveolar crystals. Histiocytic and neutrophilic pneumonia was noted in one animal that received thickened liquids. CONCLUSION: Recurrent aspiration of small amounts of thickened water resulted in significant pulmonary inflammation in a murine model of aspiration. Results of this study support the need for further investigation of liquid thickener safety and its efficacy in reducing the pulmonary complications of swallowing disorders. LEVEL OF EVIDENCE: NA Laryngoscope, 131:1223-1228, 2021.


Asunto(s)
Trastornos de Deglución/terapia , Lesión Pulmonar/inducido químicamente , Neumonía por Aspiración/prevención & control , Polisacáridos Bacterianos/farmacología , Agua/farmacología , Animales , Deglución/efectos de los fármacos , Trastornos de Deglución/complicaciones , Modelos Animales de Enfermedad , Inflamación , Pulmón/efectos de los fármacos , Neumonía por Aspiración/etiología , Estudios Prospectivos , Ratas , Ratas Sprague-Dawley , Recurrencia , Viscosidad
7.
Am J Physiol Heart Circ Physiol ; 319(6): H1162-H1180, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32986962

RESUMEN

Exosomes are a subtype of extracellular vesicles. They range from 30 to 150 nm in diameter and originate from intraluminal vesicles. Exosomes were first identified as the mechanism for releasing unnecessary molecules from reticulocytes as they matured to red blood cells. Since then, exosomes have been shown to be secreted by a broad spectrum of cells and play an important role in the cardiovascular system. Different stimuli are associated with increased exosome release and result in different exosome content. The release of harmful DNA and other molecules via exosomes has been proposed as a mechanism to maintain cellular homeostasis. Because exosomes contain parent cell-specific proteins on the membrane and in the cargo that is delivered to recipient cells, exosomes are potential diagnostic biomarkers of various types of diseases, including cardiovascular disease. As exosomes are readily taken up by other cells, stem cell-derived exosomes have been recognized as a potential cell-free regenerative therapy to repair not only the injured heart but other tissues as well. The objective of this review is to provide an overview of the biological functions of exosomes in heart disease and tissue regeneration. Therefore, state-of-the-art methods for exosome isolation and characterization, as well as approaches to assess exosome functional properties, are reviewed. Investigation of exosomes provides a new approach to the study of disease and biological processes. Exosomes provide a potential "liquid biopsy," as they are present in most, if not all, biological fluids that are released by a wide range of cell types.


Asunto(s)
Exosomas/metabolismo , Exosomas/trasplante , Insuficiencia Cardíaca/cirugía , Miocardio/patología , Regeneración , Trasplante de Células Madre , Células Madre/metabolismo , Animales , Biomarcadores/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Miocardio/metabolismo , Valor Predictivo de las Pruebas , Recuperación de la Función
8.
Cytotherapy ; 22(9): 482-485, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32425691

RESUMEN

STATEMENT: The International Society for Cellular and Gene Therapies (ISCT) and the International Society for Extracellular Vesicles (ISEV) recognize the potential of extracellular vesicles (EVs, including exosomes) from mesenchymal stromal cells (MSCs) and possibly other cell sources as treatments for COVID-19. Research and trials in this area are encouraged. However, ISEV and ISCT do not currently endorse the use of EVs or exosomes for any purpose in COVID-19, including but not limited to reducing cytokine storm, exerting regenerative effects or delivering drugs, pending the generation of appropriate manufacturing and quality control provisions, pre-clinical safety and efficacy data, rational clinical trial design and proper regulatory oversight.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas/citología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/inmunología , Exosomas/trasplante , Vesículas Extracelulares/trasplante , Humanos , Sociedades Científicas , Tratamiento Farmacológico de COVID-19
9.
J Biol Chem ; 295(12): 4001-4013, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32034092

RESUMEN

The hallmark pathological features of Alzheimer's disease (AD) brains are senile plaques, comprising ß-amyloid (Aß) peptides, and neuronal inclusions formed from tau protein. These plaques form 10-20 years before AD symptom onset, whereas robust tau pathology is more closely associated with symptoms and correlates with cognitive status. This temporal sequence of AD pathology development, coupled with repeated clinical failures of Aß-directed drugs, suggests that molecules that reduce tau inclusions have therapeutic potential. Few tau-directed drugs are presently in clinical testing, in part because of the difficulty in identifying molecules that reduce tau inclusions. We describe here two cell-based assays of tau inclusion formation that we employed to screen for compounds that inhibit tau pathology: a HEK293 cell-based tau overexpression assay, and a primary rat cortical neuron assay with physiological tau expression. Screening a collection of ∼3500 pharmaceutical compounds with the HEK293 cell tau aggregation assay, we obtained only a low number of hit compounds. Moreover, these compounds generally failed to inhibit tau inclusion formation in the cortical neuron assay. We then screened the Prestwick library of mostly approved drugs in the cortical neuron assay, leading to the identification of a greater number of tau inclusion inhibitors. These included four dopamine D2 receptor antagonists, with D2 receptors having previously been suggested to regulate tau inclusions in a Caenorhabditis elegans model. These results suggest that neurons, the cells most affected by tau pathology in AD, are very suitable for screening for tau inclusion inhibitors.


Asunto(s)
Agregado de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Antagonistas de los Receptores de Dopamina D2/química , Antagonistas de los Receptores de Dopamina D2/metabolismo , Antagonistas de los Receptores de Dopamina D2/farmacología , Células HEK293 , Humanos , Ratones , Microscopía Fluorescente , Neuronas/citología , Neuronas/metabolismo , Ratas , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Proteínas tau/antagonistas & inhibidores , Proteínas tau/genética
10.
Nucleic Acids Res ; 48(5): 2372-2387, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-31925439

RESUMEN

A significant number of X-linked genes escape from X chromosome inactivation and are associated with a distinct epigenetic signature. One epigenetic modification that strongly correlates with X-escape is reduced DNA methylation in promoter regions. Here, we created an artificial escape by editing DNA methylation on the promoter of CDKL5, a gene causative for an infantile epilepsy, from the silenced X-chromosomal allele in human neuronal-like cells. We identify that a fusion of the catalytic domain of TET1 to dCas9 targeted to the CDKL5 promoter using three guide RNAs causes significant reactivation of the inactive allele in combination with removal of methyl groups from CpG dinucleotides. Strikingly, we demonstrate that co-expression of TET1 and a VP64 transactivator have a synergistic effect on the reactivation of the inactive allele to levels >60% of the active allele. We further used a multi-omics assessment to determine potential off-targets on the transcriptome and methylome. We find that synergistic delivery of dCas9 effectors is highly selective for the target site. Our findings further elucidate a causal role for reduced DNA methylation associated with escape from X chromosome inactivation. Understanding the epigenetics associated with escape from X chromosome inactivation has potential for those suffering from X-linked disorders.


Asunto(s)
Cromosomas Humanos X/química , Epigénesis Genética , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , Inactivación del Cromosoma X , Alelos , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Dominio Catalítico , Línea Celular Tumoral , Cromosomas Humanos X/metabolismo , Islas de CpG , Edición Génica , Silenciador del Gen , Humanos , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
11.
Stem Cells ; 38(1): 15-21, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31381842

RESUMEN

Exosomes are nanovesicles secreted by virtually all cells. Exosomes mediate the horizontal transfer of various macromolecules previously believed to be cell-autonomous in nature, including nonsecretory proteins, various classes of RNA, metabolites, and lipid membrane-associated factors. Exosomes derived from mesenchymal stem/stromal cells (MSCs) appear to be particularly beneficial for enhancing recovery in various models of disease. To date, there have been more than 200 preclinical studies of exosome-based therapies in a number of different animal models. Despite a growing number of studies reporting the therapeutic properties of MSC-derived exosomes, their underlying mechanism of action, pharmacokinetics, and scalable manufacturing remain largely outstanding questions. Here, we review the global trends associated with preclinical development of MSC-derived exosome-based therapies, including immunogenicity, source of exosomes, isolation methods, biodistribution, and disease categories tested to date. Although the in vivo data assessing the therapeutic properties of MSC-exosomes published to date are promising, several outstanding questions remain to be answered that warrant further preclinical investigation.


Asunto(s)
Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Cultivadas , Humanos
12.
Stem Cells Dev ; 29(6): 327-335, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31856674

RESUMEN

Head and neck squamous cell carcinomas (HNSCCs) are malignancies that originate in the mucosal lining of the upper aerodigestive tract. Despite advances in therapeutic interventions, survival rates among HNSCC patients have remained static for years. Cancer stem cells (CSCs) are tumor-initiating cells that are highly resistant to treatment, and are hypothesized to contribute to a significant fraction of tumor recurrences. Consequently, further investigations of how CSCs mediate recurrence may provide insights into novel druggable targets. A key element of recurrence involves the tumor's ability to evade immunosurveillance. Recent published reports suggest that CSCs possess immunosuppressive properties, however, the underlying mechanism have yet to be fully elucidated. To date, most groups have focused on the role of CSC-derived secretory proteins, such as cytokines and growth factors. Here, we review the established immunoregulatory role of exosomes derived from mixed tumor cell populations, and propose further study of CSC-derived exosomes may be warranted. Such studies may yield novel insights into new druggable targets, or lay the foundation for future exosome-based diagnostics.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Exosomas/inmunología , Neoplasias de Cabeza y Cuello/inmunología , Células Madre Neoplásicas/inmunología , Carcinoma de Células Escamosas/patología , Diferenciación Celular/inmunología , Autorrenovación de las Células/inmunología , Neoplasias de Cabeza y Cuello/patología , Humanos , Recurrencia Local de Neoplasia , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología
13.
Biochem Biophys Res Commun ; 512(4): 729-735, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-30926165

RESUMEN

Mesenchymal stem cell (MSC) based therapies are currently being evaluated as a putative therapeutic in numerous human clinical trials. Recent reports have established that exosomes mediate much of the therapeutic properties of MSCs. Exosomes are nanovesicles which mediate intercellular communication, transmitting signals between cells which regulate a diverse range of biological processes. MSC-derived exosomes are packaged with numerous types of proteins and RNAs, however, their metabolomic and lipidomic profiles to date have not been well characterized. We previously reported that MSCs, in response to priming culture conditions that mimic the in vivo microenvironmental niche, substantially modulate cellular signaling and significantly increase the secretion of exosomes. Here we report that MSCs exposed to such priming conditions undergo glycolytic reprogramming, which homogenizes MSCs' metabolomic profile. In addition, we establish that exosomes derive from primed MSCs are packaged with numerous metabolites that have been directly associated with immunomodulation, including M2 macrophage polarization and regulatory T lymphocyte induction.


Asunto(s)
Exosomas/inmunología , Células Madre Mesenquimatosas/inmunología , Línea Celular , Exosomas/metabolismo , Glucólisis , Humanos , Inmunomodulación , Activación de Macrófagos , Células Madre Mesenquimatosas/metabolismo , Metaboloma , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
14.
Stem Cells Dev ; 28(6): 398-409, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30638129

RESUMEN

Mesenchymal stem cells (MSCs) facilitate functional recovery in numerous animal models of inflammatory and ischemic tissue-related diseases with a growing body of research suggesting that exosomes mediate many of these therapeutic effects. It remains unclear, however, which types of proteins are packaged into exosomes compared with the cells from which they are derived. In this study, using comprehensive proteomic analysis, we demonstrated that human primed MSCs secrete exosomes (pMEX) that are packaged with markedly higher fractions of specific protein subclasses compared with their cells of origin, indicating regulation of their contents. Notably, we found that pMEX are also packaged with substantially elevated levels of extracellular-associated proteins. Fibronectin was the most abundant protein detected, and data established that fibronectin mediates the mitogenic properties of pMEX. In addition, treatment of SHSY5Y cells with pMEX induced the secretion of growth factors known to possess mitogenic and neurotrophic properties. Taken together, our comprehensive analysis indicates that pMEX are packaged with specific protein subtypes, which may provide a molecular basis for their distinct functional properties.


Asunto(s)
Exosomas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mitosis , Adolescente , Adulto , Línea Celular Tumoral , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad
15.
J Extracell Vesicles ; 7(1): 1535750, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30637094

RESUMEN

The last decade has seen a sharp increase in the number of scientific publications describing physiological and pathological functions of extracellular vesicles (EVs), a collective term covering various subtypes of cell-released, membranous structures, called exosomes, microvesicles, microparticles, ectosomes, oncosomes, apoptotic bodies, and many other names. However, specific issues arise when working with these entities, whose size and amount often make them difficult to obtain as relatively pure preparations, and to characterize properly. The International Society for Extracellular Vesicles (ISEV) proposed Minimal Information for Studies of Extracellular Vesicles ("MISEV") guidelines for the field in 2014. We now update these "MISEV2014" guidelines based on evolution of the collective knowledge in the last four years. An important point to consider is that ascribing a specific function to EVs in general, or to subtypes of EVs, requires reporting of specific information beyond mere description of function in a crude, potentially contaminated, and heterogeneous preparation. For example, claims that exosomes are endowed with exquisite and specific activities remain difficult to support experimentally, given our still limited knowledge of their specific molecular machineries of biogenesis and release, as compared with other biophysically similar EVs. The MISEV2018 guidelines include tables and outlines of suggested protocols and steps to follow to document specific EV-associated functional activities. Finally, a checklist is provided with summaries of key points.

16.
Curr Eye Res ; 42(10): 1358-1367, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28636406

RESUMEN

PURPOSE: Exosomes derived from human mesenchymal stem cells (hMSCs) cultured under hypoxic conditions contain proteins and growth factors that promote angiogenesis. This study investigated the effect of intravitreal administration of these exosomes on retinal ischemia using a murine model. METHODS: Oxygen-induced retinopathy (OIR) was induced by exposing one-week-old male C57BL/6J mice to 5 days of 75% hyperoxic conditioning, and returning to room air. After hyperoxic conditioning, the right eye of each mouse was injected intravitreally with 1 µl saline or exosomes derived from hMSCs and compared to control mice of the same age raised in room air without OIR injected intravitreally with saline. Two weeks post-injection, fluorescein angiography (FA) and phase-variance optical coherence tomography angiography (pvOCTA) were used to assess retinal perfusion. Retinal thickness was determined by OCT. The extent of retinal neovascularization was quantitated histologically by counting vascular nuclei on the retinal surface. RESULTS: Among eyes with OIR, intravitreal exosome treatment partially preserved retinal vascular flow in vivo and reduced associated retinal thinning; retinal thickness on OCT was 111.1 ± 7.4µm with saline versus 132.1 ± 11.6µm with exosome, p < 0.001. Retinal neovascularization among OIR eyes was reduced with exosome treatment when compared to saline-treated eyes (7.75 ± 3.68 versus 2.68 ± 1.35 neovascular nuclei per section, p < 0.0001). No immunogenicity or ocular/systemic adverse effect was associated with intravitreal exosome treatment. CONCLUSIONS: Intravitreal administration of exosomes derived from hMSCs was well tolerated without immunosuppression and decreased the severity of retinal ischemia in this murine model. This appealing novel non-cellular therapeutic approach warrants further exploration.


Asunto(s)
Modelos Animales de Enfermedad , Exosomas/trasplante , Isquemia/prevención & control , Células Madre Mesenquimatosas/citología , Neovascularización Retiniana/prevención & control , Vasos Retinianos/fisiopatología , Retinopatía de la Prematuridad/prevención & control , Animales , Animales Recién Nacidos , Angiografía con Fluoresceína , Humanos , Inyecciones Intravítreas , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/toxicidad , Proteómica , Neovascularización Retiniana/diagnóstico , Neovascularización Retiniana/fisiopatología , Retinopatía de la Prematuridad/diagnóstico , Retinopatía de la Prematuridad/fisiopatología , Tomografía de Coherencia Óptica
17.
Prog Retin Eye Res ; 56: 148-165, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27784628

RESUMEN

The most common cause of untreatable vision loss is dysfunction of the retina. Conditions, such as age-related macular degeneration, diabetic retinopathy and glaucoma remain leading causes of untreatable blindness worldwide. Various stem cell approaches are being explored for treatment of retinal regeneration. The rationale for using bone marrow stem cells to treat retinal dysfunction is based on preclinical evidence showing that bone marrow stem cells can rescue degenerating and ischemic retina. These stem cells have primarily paracrine trophic effects although some cells can directly incorporate into damaged tissue. Since the paracrine trophic effects can have regenerative effects on multiple cells in the retina, the use of this cell therapy is not limited to a particular retinal condition. Autologous bone marrow-derived stem cells are being explored in early clinical trials as therapy for various retinal conditions. These bone marrow stem cells include mesenchymal stem cells, mononuclear cells and CD34+ cells. Autologous therapy requires no systemic immunosuppression or donor matching. Intravitreal delivery of CD34+ cells and mononuclear cells appears to be tolerated and is being explored since some of these cells can home into the damaged retina after intravitreal administration. The safety of intravitreal delivery of mesenchymal stem cells has not been well established. This review provides an update of the current evidence in support of the use of bone marrow stem cells as treatment for retinal dysfunction. The potential limitations and complications of using certain forms of bone marrow stem cells as therapy are discussed. Future directions of research include methods to optimize the therapeutic potential of these stem cells, non-cellular alternatives using extracellular vesicles, and in vivo high-resolution retinal imaging to detect cellular changes in the retina following cell therapy.


Asunto(s)
Células de la Médula Ósea/clasificación , Células Madre Mesenquimatosas/citología , Enfermedades de la Retina/cirugía , Trasplante de Células Madre/métodos , Animales , Humanos
18.
PLoS One ; 11(10): e0163323, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27723765

RESUMEN

In the context of the human airway, interleukin-17A (IL-17A) signaling is associated with severe inflammation, as well as protection against pathogenic infection, particularly at mucosal surfaces such as the airway. The intracellular molecule Act1 has been demonstrated to be an essential mediator of IL-17A signaling. In the cytoplasm, it serves as an adaptor protein, binding to both the intracellular domain of the IL-17 receptor as well as members of the canonical nuclear factor kappa B (NF-κB) pathway. It also has enzymatic activity, and serves as an E3 ubiquitin ligase. In the context of airway epithelial cells, we demonstrate for the first time that Act1 is also present in the nucleus, especially after IL-17A stimulation. Ectopic Act1 expression can also increase the nuclear localization of Act1. Act1 can up-regulate the expression and promoter activity of a subset of IL-17A target genes in the absence of IL-17A signaling in a manner that is dependent on its N- and C-terminal domains, but is NF-κB independent. Finally, we show that nuclear Act1 can bind to both distal and proximal promoter regions of DEFB4, one of the IL-17A responsive genes. This transcriptional regulatory activity represents a novel function for Act1. Taken together, this is the first report to describe a non-adaptor function of Act1 by directly binding to the promoter region of IL-17A responsive genes and directly regulate their transcription.


Asunto(s)
Elementos de Respuesta/fisiología , Transducción de Señal/fisiología , Transcripción Genética/fisiología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba/fisiología , Células A549 , Proteínas Adaptadoras Transductoras de Señales , Humanos , Interleucina-17/biosíntesis , Interleucina-17/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Dominios Proteicos , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , beta-Defensinas/biosíntesis , beta-Defensinas/genética
19.
Expert Opin Biol Ther ; 16(8): 1025-33, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27159050

RESUMEN

INTRODUCTION: Brain-derived neurotrophic factor (BDNF) has been implicated in wide range of neurological diseases and injury. This neurotrophic factor is vital for neuronal health, survival, and synaptic connectivity. Many therapies focus on the restoration or enhancement of BDNF following injury or disease progression. AREAS COVERED: The present review will focus on the mechanisms in which BDNF exerts its beneficial functioning, current BDNF therapies, issues and potential solutions for delivery of neurotrophic factors to the central nervous system, and other disease indications that may benefit from overexpression or restoration of BDNF. EXPERT OPINION: Due to the role of BDNF in neuronal development, maturation, and health, BDNF is implicated in numerous neurological diseases making it a prime therapeutic agent. Numerous studies have shown the therapeutic potential of BDNF in a number of neurodegenerative disease models and in acute CNS injury, however clinical translation has fallen short due to issues in delivering this molecule. The use of MSC as a delivery platform for BDNF holds great promise for clinical advancement of neurotrophic factor restoration. The ease with which MSC can be engineered opens the door to the possibility of using this cell-based delivery system to advance a BDNF therapy to the clinic.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Animales , Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Proteínas Recombinantes/uso terapéutico
20.
Cell Transplant ; 25(4): 677-86, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26850319

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by an abnormal expansion of CAG repeats. Although pathogenesis has been attributed to this polyglutamine expansion, the underlying mechanisms through which the huntingtin protein functions have yet to be elucidated. It has been suggested that postnatal reduction of mutant huntingtin through protein interference or conditional gene knockout could prove to be an effective therapy for patients suffering from HD. For allele-specific targeting, transcription activator-like effectors (TALE) were designed to target single-nucleotide polymorphisms (SNP) in the mutant allele and packaged into a vector backbone containing KRAB to promote transcriptional repression of the disease-associated allele. Additional TALEs were packaged into a vector backbone containing heterodimeric FokI and were designed to be used as nucleases (TALEN) to cause a CAG-collapse in the mutant allele. Human HD fibroblasts were treated with each TALE-SNP or TALEN. Allele-expression was measured using a SNP-genotyping assay and mutant protein aggregation was quantified with Western blots for anti-ubiquitin. The TALE-SNP and TALEN significantly reduced mutant allele expression (p < 0.05) when compared to control transfections while not affecting expression of the nondisease allele. This study demonstrates the potential of allele-specific gene modification using TALE proteins, and provides a foundation for targeted treatment for individuals suffering from Huntington's or other genetically linked diseases.


Asunto(s)
Alelos , Fibroblastos/metabolismo , Proteína Huntingtina , Enfermedad de Huntington , Polimorfismo de Nucleótido Simple , Activación Transcripcional , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...