Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Elife ; 122023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37159507

RESUMEN

Peristaltic movement of the intestine propels food down the length of the gastrointestinal tract to promote nutrient absorption. Interactions between intestinal macrophages and the enteric nervous system regulate gastrointestinal motility, yet we have an incomplete understanding of the molecular mediators of this crosstalk. Here, we identify complement component 1q (C1q) as a macrophage product that regulates gut motility. Macrophages were the predominant source of C1q in the mouse intestine and most extraintestinal tissues. Although C1q mediates the complement-mediated killing of bacteria in the bloodstream, we found that C1q was not essential for the immune defense of the intestine. Instead, C1q-expressing macrophages were located in the intestinal submucosal and myenteric plexuses where they were closely associated with enteric neurons and expressed surface markers characteristic of nerve-adjacent macrophages in other tissues. Mice with a macrophage-specific deletion of C1qa showed changes in enteric neuronal gene expression, increased neurogenic activity of peristalsis, and accelerated intestinal transit. Our findings identify C1q as a key regulator of gastrointestinal motility and provide enhanced insight into the crosstalk between macrophages and the enteric nervous system.


Asunto(s)
Complemento C1q , Sistema Nervioso Entérico , Ratones , Animales , Complemento C1q/metabolismo , Motilidad Gastrointestinal/fisiología , Macrófagos/metabolismo , Tracto Gastrointestinal
2.
Sci Rep ; 11(1): 7315, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33795767

RESUMEN

Fibrosis occurs when collagen deposition and fibroblast proliferation replace healthy tissue. Red light (RL) may improve skin fibrosis via photobiomodulation, the process by which photosensitive chromophores in cells absorb visible or near-infrared light and undergo photophysical reactions. Our previous research demonstrated that high fluence RL reduces fibroblast proliferation, collagen deposition, and migration. Despite the identification of several cellular mechanisms underpinning RL phototherapy, little is known about the transcriptional changes that lead to anti-fibrotic cellular responses. Herein, RNA sequencing was performed on human dermal fibroblasts treated with RL phototherapy. Pathway enrichment and transcription factor analysis revealed regulation of extracellular matrices, proliferation, and cellular responses to oxygen-containing compounds following RL phototherapy. Specifically, RL phototherapy increased the expression of MMP1, which codes for matrix metalloproteinase-1 (MMP-1) and is responsible for remodeling extracellular collagen. Differential regulation of MMP1 was confirmed with RT-qPCR and ELISA. Additionally, RL upregulated PRSS35, which has not been previously associated with skin activity, but has known anti-fibrotic functions. Our results suggest that RL may benefit patients by altering fibrotic gene expression.


Asunto(s)
Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Fototerapia/métodos , Piel/metabolismo , Piel/efectos de la radiación , Transcriptoma , Adulto , Movimiento Celular , Proliferación Celular , Colágeno/metabolismo , Femenino , Fibrosis , Perfilación de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 1 de la Matriz/biosíntesis , Persona de Mediana Edad , Estrés Oxidativo , Oxígeno/metabolismo , RNA-Seq , Especies Reactivas de Oxígeno , Enfermedades de la Piel/metabolismo , Factores de Tiempo , Factores de Transcripción
3.
J Biophotonics ; 12(5): e201800207, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30182520

RESUMEN

Skin fibrosis is a chronic debilitating feature of several skin diseases that lead to characteristic increases in dermal fibroblast proliferation and collagen deposition through upregulation in components of the transforming growth factor beta (TGF-B)/SMAD pathway. In contrast to ultraviolet phototherapy, high-fluence light-emitting diode-generated red light (HF-LED-RL, 633 ± 15 nm) is a safe, economic and non-invasive therapy with in vitro evidence that supports modulation of the key cellular characteristics involved in the pathogenesis of skin fibrosis. Limited data exists pertaining to the effects of HF-LED-RL on human skin fibroblast microRNA (miRNA). Herein, we explored the effects of HF-LED-RL on fibroblast miRNA levels using RNA-seq and miRNA expression analysis. Using RNA-seq analysis we found that HF-LED-RL at 320 and 640 J/cm2 increased transcription of key miRNA that are involved in skin fibrosis including miRNA-29, miRNA-196a and Let-7a, and decreased transcription of miRNA-21, miRNA-23b and miRNA-31. These microRNA findings provide insight into the molecular underpinnings of HF-LED-RL and highlight potential therapeutic targets of interest for the treatment of skin fibrosis. Additional research on the specific molecular mechanisms underlying HF-LED-RL effects on fibroblasts may provide further mechanistic insight into this therapy and may reveal additional future therapeutic targets for skin fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Luz , MicroARNs/genética , Piel/citología , Transcriptoma/efectos de la radiación , Humanos , Fototerapia
4.
Sci Rep ; 8(1): 12599, 2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30135507

RESUMEN

Thermal photodynamic therapy (PDT) is an emerging modality to optimize treatment of pre-cancerous squamous cell carcinoma (SCC) lesions, known as actinic keratoses. Thermal PDT involves heating the tissue, skin, or mucosa above normal skin temperature during 5-aminolevulinic (5-ALA) incubation and irradiating with blue light, which leads to cell apoptosis and reactive oxygen species (ROS) generation. To our knowledge, thermal PDT has not been studied for the treatment of cutaneous or mucosal SCC. We incubated two SCC cell lines with 5-ALA for 30 minutes at temperatures between 21 °C and 42 °C and then irradiated cells with 1000 seconds of blue light. We measured changes in apoptosis, necrosis, and ROS. At 36 °C, there was a dose-dependent increase in apoptosis and ROS generation. Thermal incubation of 5-ALA at 39° and 42 °C followed by blue light increased cell apoptosis and ROS generation compared to untreated control samples incubated at the same temperatures. Thermal PDT may represent a new treatment option for cutaneous and mucosal SCC cancer. Thermal PDT is associated with an increase in SCC cellular apoptosis and is associated with an upregulation in ROS. Clinical trials are required to determine optimal thermal PDT treatment parameters and efficacy for cutaneous and mucosal SCC.


Asunto(s)
Calor/uso terapéutico , Queratosis Actínica/terapia , Fotoquimioterapia/métodos , Ácido Aminolevulínico/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/terapia , Línea Celular Tumoral , Humanos , Queratosis Actínica/metabolismo , Membrana Mucosa/metabolismo , Fármacos Fotosensibilizantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Piel/metabolismo
5.
Dermatol Surg ; 44(2): 149-157, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29401161

RESUMEN

BACKGROUND: Keloids and hypertrophic scars are conditions of pathologic scarring characterized by fibroblast hyperproliferation and excess collagen deposition. These conditions significantly impact patients by causing psychosocial, functional, and aesthetic distress. Current treatment modalities have limitations. Clinical evidence indicates that botulinum toxin A (BoNT-A) may prevent and treat keloids and hypertrophic scars. OBJECTIVE: To examine investigated cellular pathways involved in BoNT-A therapeutic modulation of keloids and hypertrophic scars. METHODS: The authors searched PubMed, Embase, and Web of Science for basic science articles related to botulinum toxin therapy, scarring, fibroblasts, keloids, and hypertrophic scars. RESULTS: Eleven basic science articles involving keloids and hypertrophic scars were reviewed. DISCUSSION: BoNT-A may reduce skin fibrosis by decreasing fibroblast proliferation, modulating the activity of transforming growth factor-ß, and reducing transcription and expression of profibrotic cytokines in keloid-derived and hypertrophic scar-derived dermal fibroblasts. BoNT-A may modulate collagen deposition, but there is a paucity of evidence regarding specific mechanisms of action. CONCLUSION: Overall, BoNT-A has the potential to prevent or treat pathologic scars in patients with a known personal or family history of keloids and hypertrophic scars, which may improve patient psychosocial distress and reduce clinic visits and health care costs. Variability in keloid and hypertrophic scar response to BoNT-A may be due to interexperiment differences in dosing, tissue donors, and assay sensitivity.


Asunto(s)
Toxinas Botulínicas Tipo A/uso terapéutico , Cicatriz Hipertrófica/tratamiento farmacológico , Queloide/tratamiento farmacológico , Fármacos Neuromusculares/uso terapéutico , Humanos
6.
Dermatol Surg ; 44(4): 528-533, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29053537

RESUMEN

BACKGROUND: Actinic keratoses (AKs) prevalence was estimated at 39.5 million Americans in 2004, and the cost to treat AKs that year was approximately 1 billion dollars. Photodynamic therapy (PDT) is an FDA-approved therapy for the treatment of AK. Recent studies have focused on reducing PDT treatment time while maintaining efficacy. OBJECTIVE: To investigate the use of thermal modulation to improve the efficacy of ultra short aminolevulinic acid (ALA) incubation PDT. MATERIALS AND METHODS: Human dermal fibroblasts (HDFs) were incubated for 10, 15, or 20 minutes with 0.5-mM ALA at various temperatures (21, 24, 27, 30, 33, 36, 39, and 42°C). After ALA incubation, samples were treated for 1,000 seconds with blue light (417 ± 5 nm) resulting in a fluence of 10 J/cm. Samples were collected and stained for apoptosis/necrosis with annexin-V and 7-aminoactinomycin D (7-AAD), then analyzed by flow cytometry. RESULTS: Human dermal fibroblast treated with 10-minute ALA-PDT had no statistically significant changes in apoptosis at all temperatures. Human dermal fibroblast treated with 15- or 20-minute ALA-PDT had statistically significant increases in apoptosis at 39 and 42°C (p < .05). CONCLUSION: These results suggest the use of thermal modulation may improve ultra short ALA incubation PDT efficacy.


Asunto(s)
Ácido Aminolevulínico/farmacología , Muerte Celular/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Técnicas de Cultivo de Célula , Muerte Celular/efectos de los fármacos , Fibroblastos/patología , Humanos
7.
J Drugs Dermatol ; 16(5): 482-488, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28628685

RESUMEN

Background: Psoriasis is a chronic, inflammatory skin condition. The economic burden of psoriasis is approximately $35.2 billion in the United States per year, and treatment costs are increasing at a higher rate than general inflation. Light emitting diode (LED) phototherapy may represent a cost-effective, efficacious, safe, and portable treatment modality for psoriasis.

Objective: The goal of our manuscript is to review the published literature and provide evidence-based recommendations on LED phototherapy for the treatment of psoriasis.

Methods & Materials: A search of the databases Pubmed, EMBASE, Web of Science, and CINAHL was performed on April 5, 2016. Key search terms were related to psoriasis and LED-based therapies.

Results: A total of 7,793 articles were generated from the initial search and 5 original articles met inclusion criteria for our review. Grade of recommendation: B for LED-blue light. Grade of recommendation: C for LED-ultraviolet B, LED-red light, and combination LED-near-infrared and LED-red light.

Conclusion: We envision further characterizing the effects of LED phototherapy to treat psoriasis in patients may increase adoption of LED-based modalities and provide clinicians and patients with new therapeutic options that balance safety, efficacy, and cost.

J Drugs Dermatol. 2017;16(5):482-488.

.


Asunto(s)
Fototerapia/métodos , Fototerapia/tendencias , Psoriasis/radioterapia , Análisis Costo-Beneficio/tendencias , Humanos , Fototerapia/economía , Psoriasis/diagnóstico , Psoriasis/economía , Resultado del Tratamiento , Terapia Ultravioleta/economía , Terapia Ultravioleta/métodos , Terapia Ultravioleta/tendencias
8.
Lasers Surg Med ; 49(6): 592-598, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28370019

RESUMEN

BACKGROUND AND OBJECTIVE: The estimated incidence of cutaneous squamous cell carcinoma (SCC) is 700,000 cases per year. In the US, SCC incidence is highest among fair skinned adults older than 50 years of age. Thus, as the population ages, the reported number of SCCs will likely increase in the future. Photodynamic therapy (PDT) is an FDA approved therapy for treatment of actinic keratoses (AKs), a precursor to cutaneous SCC lesions. The FDA approved incubation time of the photosensitizing agent 5-aminolevulinic acid (ALA) is 14-18 hours. Recent studies have investigated short ALA incubation times of 1-3 hours with ALA and PDT demonstrating treatment success. Therefore, the question exists whether ALA incubation periods of less than 30 minutes are efficacious. Herein, we evaluate the efficacy of short ALA incubation periods by measuring apoptosis after 10, 15, and 20 minutes of ALA incubation. STUDY DESIGN/MATERIALS AND METHODS: AG13145 normal human dermal fibroblasts HDFs were incubated with 10, 15, or 20 minute of ALA at various concentrations (0, 0.05, 0.075, 0.1, 0.25, 0.375, 0.5, 1, and 2 mM). After ALA incubation, samples were treated with 1,000 seconds (16 minutes 40 seconds) of Blu-U fluorescent blue light (417 ± 5 nm) for a fluence of 10 J/cm2 . Immediately following treatment with blue light, samples were collected and stained for apoptosis and necrosis with annexin-V and 7-aminoactinomycin D (7-AAD), and then analyzed by flow cytometry. RESULTS: HDFs incubated with ALA for 10 minute at 36 °C followed by 10 J/cm2 of blue light had no statistically significant changes in apoptosis. HDFs incubated with ALA for 15 or 20 minutes at 36 °C followed by 10 J/cm2 of blue light had statistically significant increases in the percentages of cells positive for apoptosis in the 0.5, 1.0, and 2.0 mM ALA doses (P < 0.05). CONCLUSIONS: We found that incubation of ALA for at least 15 minutes followed by 10 J/cm2 of blue light resulted in a statistically significant increase in apoptosis. Lasers Surg. Med. 49:592-598, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Ácido Aminolevulínico/farmacología , Apoptosis/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Ácido Aminolevulínico/uso terapéutico , Células Cultivadas , Esquema de Medicación , Fibroblastos/patología , Humanos , Queratosis Actínica/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Factores de Tiempo
9.
J Drugs Dermatol ; 15(7): 843-8, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27391634

RESUMEN

BACKGROUND: Diabetic foot ulcers (DFU) represent a significant complication of diabetes mellitus (DM). DFU affect one in four patients with DM and treatments of DFU are limited and challenging. The management of DFU remains a significant healthcare and socioeconomic burden ($245 billion). There is a wide range of advanced therapies for DFU, but these are costly and have demonstrated only minimal efficacy in limited published studies. An emerging treatment modality to improve DFU and optimize wound healing is the use of low-level light therapy (LLLT). LLLT involves the use of light in the form of low-level or low-power laser or light emitting diodes to alter biochemical pathways, which may result in changes to cell shape, cell migration, and cell signaling.
OBJECTIVE: To review published clinical experiences (case series and case reports) using LLLT for treatment of DFU, and provide evidence-based recommendations and future directions on the potential of LLLT as a therapeutic modality for DFU.
METHODS AND MATERIALS: On January 16, 2016 we searched the published literature using databases: PubMed, EMBASE, CINAHL, and Web of Science with key terms: "diabetic foot" AND ("low level laser therapy" OR "low level light therapy" OR "LLLT" OR "light emitting diode" OR "phototherapy" OR "laser").
RESULTS: After screening of titles, abstracts and/or full-text, 7 original articles were suitable in our review. Our review contains 5 case series and 2 case reports that evaluated LLLT for treatment of DFU, and all reviewed studies have shown positive improvement of DFU using LLLT with no adverse events, albeit with limitations that may be minimized with future RCTs.
CONCLUSIONS: LLLT is an emerging and promising treatment modality to current alternatives that are costly and have shown limited success. Based upon the published evidence, we envision additional research may allow for stronger recommendation with LLLT for treatment of DFU.

J Drugs Dermatol. 2016;15(7):843-848.


Asunto(s)
Diabetes Mellitus/diagnóstico , Diabetes Mellitus/radioterapia , Pie Diabético/diagnóstico , Pie Diabético/radioterapia , Terapia por Luz de Baja Intensidad/métodos , Humanos , Resultado del Tratamiento
10.
J Biophotonics ; 9(11-12): 1167-1179, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27174640

RESUMEN

Skin fibrosis, often referred to as skin scarring, is a significant international health problem with limited treatment options. The hallmarks of skin fibrosis are increased fibroblast proliferation, collagen production, and migration speed. Recently published clinical observations indicate that visible red light may improve skin fibrosis. In this study we hypothesize that high-fluence light-emitting diode-generated red light (HF-LED-RL) modulates the key cellular features of skin fibrosis by decreasing cellular proliferation, collagen production, and migration speed of human skin fibroblasts. Herein, we demonstrate that HF-LED-RL increases reactive oxygen species (ROS) generation for up to 4 hours, inhibits fibroblast proliferation without increasing apoptosis, inhibits collagen production, and inhibits migration speed through modulation of the phosphoinositide 3-kinase (PI3K)/Akt pathway. We demonstrate that HF-LED-RL is capable of inhibiting the unifying cellular processes involved in skin fibrosis including fibroblast proliferation, collagen production, and migration speed. These findings suggest that HF-LED-RL may represent a new approach to treat skin fibrosis. LED advantages include low cost, portability, and ease of use. Further characterizing the photobiomodulatory effects of HF-LED-RL on fibroblasts and investigating the anti-fibrotic effects of HF-LED-RL in human subjects may provide new insight into the utility of this therapeutic approach for skin fibrosis.


Asunto(s)
Fibroblastos/efectos de la radiación , Terapia por Luz de Baja Intensidad , Piel/patología , Proliferación Celular/efectos de la radiación , Células Cultivadas , Colágeno/biosíntesis , Fibroblastos/patología , Fibrosis , Humanos , Luz , Fosfatidilinositol 3-Quinasas/metabolismo , Piel/efectos de la radiación
11.
Dermatol Surg ; 42(6): 727-32, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27176863

RESUMEN

BACKGROUND: Light-emitting diode-generated blue light (LED-BL) is part of the visible light spectrum that does not cause DNA damage and may represent a safer alternative to ultraviolet phototherapy. Previous research demonstrated that LED-BL can inhibit adult human skin fibroblast proliferation and migration speed and is associated with increased reactive oxygen species (ROS) generation in a dose-dependent manner. In addition, resveratrol possesses potent intracellular antioxidative effects on ROS-free radicals in human skin fibroblasts. OBJECTIVE: The authors studied the effects on migration speed as a surrogate to measure LED-BL effects on fibroblast function. The authors hypothesized that resveratrol, a potent scavenger of ROS, could prevent the effects of LED-BL on fibroblast migration speed. This would implicate ROS as the mechanistic driver of LED-BL effects on human skin fibroblasts. METHODS: To demonstrate that resveratrol could prevent the effects of LED-BL (415-nm), fibroblasts were incubated with resveratrol (Sigma-Aldrich, St. Louis, MO) at concentrations of 0.001% and 0.0001% for 24 hours and then irradiated with LED-BL at fluences of 30, 45, and 80 J/cm. Postirradiation fibroblast migratory speed was assayed in an environment-controlled computer-assisted video microscopy system. Reactive oxygen species levels were measured by flow cytometric analysis of dihydrorhodamine. Statistical analyses with analysis of variance and Student t-test were performed to compare individual treatment arms and matched controls. RESULTS: The experimental results demonstrate that pretreatment of skin fibroblasts with resveratrol at concentrations of 0.001% and 0.0001% prevents the effects of 30, 45, and 80 J/cm of LED-BL on fibroblast migration speed. The authors found that LED-BL at a fluences of 30, 45, and 80 J/cm significantly increased ROS, whereas pretreatment with 0.001% resveratrol significantly reduced ROS generation. CONCLUSION: The findings demonstrate that LED-BL-induced decreases in fibroblast migration speed can be prevented by pretreating cells with resveratrol. This finding supports the hypothesis that ROS generation is the most likely driver of LED-BL-induced alterations in migration speed and suggests that ROS generation may be responsible for a number of other alterations seen after LED-BL phototherapy, such as decreases in cellular migration, cytokine levels, and myofibroblast differentiation. The authors hypothesize that their findings may result in greater understanding of the fundamental mechanisms underlying visible light interaction with skin and they hope dermatologists and other researchers may use these pathways for patient benefit.


Asunto(s)
Antioxidantes/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Luz , Estilbenos/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Células Cultivadas , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Citometría de Flujo , Humanos , Especies Reactivas de Oxígeno/efectos de la radiación , Resveratrol , Piel/citología
12.
Wound Repair Regen ; 24(2): 418-26, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-26748691

RESUMEN

Diabetes mellitus (DM) is a significant international health concern affecting more than 387 million individuals. A diabetic person has a 25% lifetime risk of developing a diabetic foot ulcer (DFU), leading to limb amputation in up to one in six DFU patients. Low-level light therapy (LLLT) uses low-power lasers or light-emitting diodes to alter cellular function and molecular pathways, and may be a promising treatment for DFU. The goal of this systematic review is to examine whether the clinical use of LLLT is effective in the healing of DFU at 12 and 20 weeks in comparison with the standard of care, and to provide evidence-based recommendation and future clinical guidelines for the treatment of DFU using LLLT. On September 30, 2015, we searched PubMed, EMBASE, CINAHL, and Web of Science databases using the following terms: "diabetic foot" AND "low level light therapy," OR "light emitting diode," OR "phototherapy," OR "laser." The relevant articles that met the following criteria were selected for inclusion: randomized control trials (RCTs) that investigated the use of LLLT for treatment of DFU. Four RCTs involving 131 participants were suitable for inclusion based upon our criteria. The clinical trials used sham irriadiation, low dose, or nontherapeutic LLLT as placebo or control in comparison to LLLT. The endpoints included ulcer size and time to complete healing with follow-up ranging from 2 to 16 weeks. Each article was assigned a level of evidence (LOE) and graded according to the Oxford Center for Evidence-based Medicine Levels of Evidence Grades of Recommendation criteria. Limitations of reviewed RCTs include a small sample size (N < 100), unclear allocation concealment, lack of screening phase to exclude rapid healers, unclear inclusion/exclusion criteria, short (<30 days) follow-up period, and unclear treatment settings (wavelength and treatment time). However, all reviewed RCTs demonstrated therapeutic outcomes with no adverse events using LLLT for treatment of DFU. This systematic review reports that LLLT has significant potential to become a portable, minimally invasive, easy-to-use, and cost effective modality for treatment of DFU. To enthusiastically recommend LLLT for treatment of DFU, additional studies with comparable laser parameters, screening period to exclude rapid healers, larger sample sizes and longer follow-up periods are required. We envision future stringent RCTs may validate LLLT for treatment of DFU. Systematic review registration number: PROSPERO CRD42015029825.


Asunto(s)
Pie Diabético/terapia , Terapia por Luz de Baja Intensidad , Cicatrización de Heridas/fisiología , Amputación Quirúrgica/estadística & datos numéricos , Análisis Costo-Beneficio , Pie Diabético/patología , Medicina Basada en la Evidencia , Humanos , Terapia por Luz de Baja Intensidad/economía , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
13.
PLoS One ; 10(10): e0140628, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26488596

RESUMEN

BACKGROUND: Skin fibrosis is a significant medical problem that leads to a functional, aesthetic, and psychosocial impact on quality-of-life. Light-emitting diode-generated 633-nm red light (LED-RL) is part of the visible light spectrum that is not known to cause DNA damage and is considered a safe, non-invasive, inexpensive, and portable potential alternative to ultraviolet phototherapy that may change the treatment paradigm of fibrotic skin disease. OBJECTIVE: The goal of our study was to investigate the how reactive oxygen species (ROS) free radicals generated by high fluence LED-RL inhibit the migration of skin fibroblasts, the main cell type involved in skin fibrosis. Fibroblast migration speed is increased in skin fibrosis, and we studied cellular migration speed of cultured human skin fibroblasts as a surrogate measure of high fluence LED-RL effect on fibroblast function. To ascertain the inhibitory role of LED-RL generated ROS on migration speed, we hypothesized that resveratrol, a potent antioxidant, could prevent the photoinhibitory effects of high fluence LED-RL on fibroblast migration speed. METHODS: High fluence LED-RL generated ROS were measured by flow cytometry analysis using dihydrorhodamine (DHR). For purposes of comparison, we assessed the effects of ROS generated by hydrogen peroxide (H2O2) on fibroblast migration speed and the ability of resveratrol, a well known antioxidant, to prevent LED-RL and H2O2 generated ROS-associated changes in fibroblast migration speed. To determine whether resveratrol could prevent the high fluence LED-RL ROS-mediated photoinhibition of human skin fibroblast migration, treated cells were incubated with resveratrol at concentrations of 0.0001% and 0.001% for 24 hours, irradiated with high fluences LED-RL of 480, 640, and 800 J/cm2. RESULTS: High fluence LED-RL increases intracellular fibroblast ROS and decreases fibroblast migration speed. LED-RL at 480, 640 and 800 J/cm2 increased ROS levels to 132.8%, 151.0%, and 158.4% relative to matched controls, respectively. These LED-RL associated increases in ROS were prevented by pretreating cells with 0.0001% or 0.001% resveratrol. Next, we quantified the effect of hydrogen peroxide (H2O2)-associated ROS on fibroblast migration speed, and found that while H2O2-associated ROS significantly decreased relative fibroblast migration speed, pretreatment with 0.0001% or 0.001% resveratrol significantly prevented the decreases in migration speed. Furthermore, we found that LED-RL at 480, 640 and 800 J/cm2 decreased fibroblast migration speed to 83.0%, 74.4%, and 68.6% relative to matched controls, respectively. We hypothesized that these decreases in fibroblast migration speed were due to associated increases in ROS generation. Pretreatment with 0.0001% and 0.001% resveratrol prevented the LED-RL associated decreases in migration speed. CONCLUSION: High fluence LED-RL increases ROS and is associated with decreased fibroblast migration speed. We provide mechanistic support that the decreased migration speed associated with high fluence LED-RL is mediated by ROS, by demonstrating that resveratrol prevents high fluence LED-RL associated migration speed change. These data lend support to an increasing scientific body of evidence that high fluence LED-RL has anti-fibrotic properties. We hypothesize that our findings may result in a greater understanding of the fundamental mechanisms underlying visible light interaction with skin and we anticipate clinicians and other researchers may utilize these pathways for patient benefit.


Asunto(s)
Antioxidantes/farmacología , Movimiento Celular/fisiología , Fibroblastos/metabolismo , Peróxido de Hidrógeno/farmacología , Estilbenos/farmacología , Proliferación Celular/fisiología , Células Cultivadas , Fibrosis , Humanos , Luz , Fototerapia , Resveratrol , Piel/fisiopatología
14.
Mol Cell Endocrinol ; 203(1-2): 65-75, 2003 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-12782404

RESUMEN

Autoregulation of thyroid hormone (TH) receptors (TRs) is a mechanism whereby a cell can regulate its responsiveness to TH. Nuclear coactivators (NCoAs) modulate TH action and may also be important for regulation of TR expression. We have determined the effect of TH withdrawal and treatment on the expression of different isoforms of TR as well as expression of the NCoAs SRC-1, TIF-2 and SRC-3 using quantitative real time polymerase chain reaction. In order to identify the effect that each TR isoform exerts over the expression of the other, NCoA and TR transcripts were measured in liver and heart tissue from wild type mice or mice with deletion of either TR isoform or SRC-1 genes. In liver, regulation of TR beta1 and TR alpha2 subtype expression is inversely related to TH levels and the regulation of TR beta expression is, in part, controlled by TR alpha. In the heart, the opposite is the case, regulation of TR alpha2 and TR beta1 isoform expression is directly related to TH levels and this regulation is primarily controlled by TR alpha. Although NCoAs are, in general, increased in response to hypothyroidism or in states of TH resistance, SRC-1 specifically does not regulate TR isoform expression. We have demonstrated that TR isoforms and NCoAs are autoregulated transcription factors with tissue specificity.


Asunto(s)
Regulación de la Expresión Génica , Receptores de Hormona Tiroidea/genética , Hormonas Tiroideas/fisiología , Factores de Transcripción/genética , Animales , Histona Acetiltransferasas , Hígado/metabolismo , Ratones , Ratones Noqueados , Miocardio/metabolismo , Coactivador 1 de Receptor Nuclear , Coactivador 2 del Receptor Nuclear , Coactivador 3 de Receptor Nuclear , Isoformas de Proteínas/genética , ARN Mensajero/análisis , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/genética , Transactivadores/genética
15.
Mol Endocrinol ; 17(5): 882-94, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12576486

RESUMEN

Steroid receptor coactivator-1 (SRC-1) is a transcription cofactor that enhances the hormone-dependent action mediated by the thyroid hormone (TH) receptor (TR) as well as other nuclear receptors. However, it is not known whether the SRC-1-mediated activation of TH-regulated gene transcription is TR isoform specific in the pituitary. We generated mice that were deficient in TRalpha and SRC-1 (TRalpha(0/0)SRC-1(-/-)), as well in TRbeta and SRC-1 (TRbeta(-/-)SRC-1(-/-)), and thyroid function tests and effects of TH deprivation and TH treatment were compared with wild-type mice or mice with deletion of either TRs or SRC-1 alone. We have shown that 1) TRbeta(-/-)SRC-1(-/-) mice demonstrate more severe TH resistance than either the SRC-1(-/-) or TRbeta(-/-) mice; the additive effect indicates that SRC-1 has an independent role in TH action over that of TRbeta; 2) SRC-1 facilitates TRbeta and TRalpha-mediated down-regulation of TSH, as TRalpha(0/0)SRC-1(-/-) mice demonstrate TH resistance rather than hypersensitivity as seen in TRalpha(0/0)mice; and 3) a compensatory increase in SRC-1 expression is associated with the TH hypersensitivity seen in TRalpha-deficient animals. We conclude that SRC-1 action in the pituitary mediates TH action via specific TR subtypes.


Asunto(s)
Hipófisis/fisiología , Receptores de Hormona Tiroidea/metabolismo , Receptores alfa de Hormona Tiroidea/metabolismo , Factores de Transcripción/metabolismo , Animales , Núcleo Celular/metabolismo , Femenino , Histona Acetiltransferasas , Masculino , Ratones , Ratones Mutantes , Coactivador 1 de Receptor Nuclear , Coactivador 2 del Receptor Nuclear , Coactivador 3 de Receptor Nuclear , Hipófisis/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Receptores de Hormona Tiroidea/genética , Pruebas de Función de la Tiroides , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea , Síndrome de Resistencia a Hormonas Tiroideas/tratamiento farmacológico , Síndrome de Resistencia a Hormonas Tiroideas/genética , Síndrome de Resistencia a Hormonas Tiroideas/metabolismo , Hormonas Tiroideas/deficiencia , Hormonas Tiroideas/metabolismo , Hormonas Tiroideas/farmacología , Tirotropina/sangre , Tirotropina/genética , Tirotropina/metabolismo , Tirotropina de Subunidad beta/efectos de los fármacos , Tirotropina de Subunidad beta/genética , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Triyodotironina/metabolismo
16.
Am J Physiol Endocrinol Metab ; 283(3): E428-35, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12169435

RESUMEN

We investigated the effect of thyroid hormone (TH) receptor (TR)alpha and -beta isoforms in TH action in the heart. Noninvasive echocardiographic measurements were made in mice homozygous for disruption of TRalpha (TRalpha(0/0)) or TRbeta (TRbeta(-/-)). Mice were studied at baseline, 4 wk after TH deprivation (using a low-iodine diet containing propylthiouracil), and after 4-wk treatment with TH. Baseline heart rates (HR) were similar in wild-type (WT) and TRalpha(0/0) mice but were greater in TRbeta(-/-) mice. With TH deprivation, HR decreased 49% in WT and 37% in TRbeta(-/-) mice and decreased only 5% in TRalpha(0/0) mice from baseline, whereas HR increased in all genotypes with TH treatment. Cardiac output (CO) and cardiac index (CI) in WT mice decreased (-31 and -32%, respectively) with TH deprivation and increased (+69 and +35%, respectively) with TH treatment. The effects of CO and CI were blunted with TH withdrawal in both TRalpha(0/0) (+8 and -2%, respectively) and TRbeta(-/-) mice (-17 and -18%, respectively). Treatment with TH resulted in a 64% increase in LV mass in WT and a 44% increase in TRalpha(0/0) mice but only a 6% increase in TRbeta(-/-) mice (ANOVA P < 0.05). Taken together, these data suggest that TRalpha and TRbeta play different roles in the physiology of TH action on the heart.


Asunto(s)
Corazón/fisiología , Receptores de Hormona Tiroidea/fisiología , Hormonas Tiroideas/fisiología , Animales , Peso Corporal , Ecocardiografía , Frecuencia Cardíaca/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/genética , Receptores de Hormona Tiroidea/deficiencia , Receptores de Hormona Tiroidea/genética , Pruebas de Función de la Tiroides , Receptores beta de Hormona Tiroidea
17.
J Clin Invest ; 109(4): 469-73, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11854318

RESUMEN

Three genes, TTF1, TTF2, and PAX8, involved in thyroid gland development and migration have been identified. Yet systematic screening for defects in these genes in thyroid dysgenesis gave essentially negative results. In particular, no TTF1 gene defects were found in 76 individuals with thyroid dysgenesis even though a deletion of this gene in the mouse results in thyroid and lung agenesis and defective diencephalon. We report a 6-year-old boy with predominant dyskinesia, neonatal respiratory distress, and mild hyperthyrotropinemia. One allele of his TTF1 gene had a guanidine inserted into codon 86 producing a nonsense protein of 407, rather than 371, amino acids. The mutant TTF1 did not bind to its canonical cis-element or transactivate a reporter gene driven by the thyroglobulin promoter, a natural target of TTF1. Failure of the mutant TTF1 to interfere with binding and transactivation functions of the wild-type TTF1 suggested that the syndrome was caused by haploinsufficiency. This was confirmed in mice heterozygous for Ttf1 gene deletion, heretofore considered to be normal. Compared with wild-type littermates, Ttf1(+/-) mice had poor coordination and a significant elevation of serum thyrotropin. Therefore, haploinsufficiency of the TTF1 gene results in a predominantly neurological phenotype and secondary hyperthyrotropinemia.


Asunto(s)
Enfermedades del Sistema Nervioso Central/etiología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Glándula Tiroides/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/metabolismo , Niño , ADN/genética , Análisis Mutacional de ADN , Femenino , Humanos , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/metabolismo , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación , Fenotipo , Síndrome , Enfermedades de la Tiroides/etiología , Enfermedades de la Tiroides/genética , Enfermedades de la Tiroides/metabolismo , Factor Nuclear Tiroideo 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...