Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.849
Filtrar
1.
An Acad Bras Cienc ; 96(suppl 1): e20231139, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39140521

RESUMEN

Tendons, complex fibrous structures, are subjected to great tensions, which can give rise to the so-called tendinopathies. This study aimed to evaluate photobiomodulation and human Amniotic Membrane applied as single or combined therapies to treat induced Achilles tendon lesions. Seventy-five rats were divided into five groups (n=15): C- control Sham surgery; I- tendon injury; LA- tendon injury treated with photobiomodulation; AM- tendon injury treated with Amniotic Membrane; LAM- tendon injury + photobiomodulation and Amniotic Membrane, subdivided into three groups (n=5) with analysis at 3, 7, and 14 days. The tendon injuries were made with a 20 g weight released from a mini guillotine onto the ankle in dorsiflexion. AM and LAM groups received an Amniotic Membrane fragment while LA and LAM groups received transcutaneous photobiomodulation, using a 660 nm wavelength laser. The inflammatory cells showed statistical differences between groups C and I (p<0.05), I and AM (p<0.01), I and LA (p<0.05), and I and LAM (p<0.01). Both photobiomodulation and Amniotic Membrane were shown to enhance tendon repair, and the association of photobiomodulation plus Amniotic Membrane was the most effective treatment. We conclude that the association of photobiomodulation plus Amniotic Membrane was effective in accelerating and improving the tendon regeneration process.


Asunto(s)
Tendón Calcáneo , Amnios , Terapia por Luz de Baja Intensidad , Ratas Wistar , Traumatismos de los Tendones , Animales , Terapia por Luz de Baja Intensidad/métodos , Amnios/trasplante , Amnios/efectos de la radiación , Traumatismos de los Tendones/terapia , Traumatismos de los Tendones/radioterapia , Tendón Calcáneo/lesiones , Tendón Calcáneo/efectos de la radiación , Ratas , Cicatrización de Heridas/efectos de la radiación , Cicatrización de Heridas/fisiología , Masculino , Humanos , Modelos Animales de Enfermedad
2.
J Wound Care ; 33(8): 612-616, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39140406

RESUMEN

There are several reasons for skin damage, including genetic factors, disorders, acute trauma, hard-to-heal wounds, or surgical interventions. Whatever the cause, wounds have a substantial impact on people who experience them, their caregivers and the healthcare system. Advanced wound care products have been researched and developed, providing an opportunity for faster and more complete healing. Tissue engineering (TE) is a promising strategy that can overcome limitations when choosing a graft for a wound. Amniotic membrane is a highly abundant, readily available, and inexpensive biological tissue that does not raise ethical concerns, with many applications in different fields of TE and regenerative medicine. It has attractive physical characteristics, such as elasticity, rigidity and mechanical strength, among others. The effects can also be potentiated by association with other substances, such as hyaluronic acid and growth factors. This paper describes new perspectives involving the use of amniotic membranes.


Asunto(s)
Amnios , Ingeniería de Tejidos , Cicatrización de Heridas , Humanos , Amnios/trasplante , Heridas y Lesiones/terapia , Medicina Regenerativa/métodos
3.
PLoS One ; 19(8): e0309063, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39159152

RESUMEN

During pregnancy, two fetomaternal interfaces, the placenta-decidua basalis and the fetal membrane-decidua parietals, allow for fetal growth and maturation and fetal-maternal crosstalk, and protect the fetus from infectious and inflammatory signaling that could lead to adverse pregnancy outcomes. While the placenta has been studied extensively, the fetal membranes have been understudied, even though they play critical roles in pregnancy maintenance and the initiation of term or preterm parturition. Fetal membrane dysfunction has been associated with spontaneous preterm birth (PTB, < 37 weeks gestation) and preterm prelabor rupture of the membranes (PPROM), which is a disease of the fetal membranes. However, it is unknown how the individual layers of the fetal membrane decidual interface (the amnion epithelium [AEC], the amnion mesenchyme [AMC], the chorion [CTC], and the decidua [DEC]) contribute to these pregnancy outcomes. In this study, we used a single-cell transcriptomics approach to unravel the transcriptomics network at spatial levels to discern the contributions of each layer of the fetal membranes and the adjoining maternal decidua during the following conditions: scheduled caesarian section (term not in labor [TNIL]; n = 4), vaginal term in labor (TIL; n = 3), preterm labor with and without rupture of membranes (PPROM; n = 3; and PTB; n = 3). The data included 18,815 genes from 13 patients (including TIL, PTB, PPROM, and TNIL) expressed across the four layers. After quality control, there were 11,921 genes and 44 samples. The data were processed by two pipelines: one by hierarchical clustering the combined cases and the other to evaluate heterogeneity within the cases. Our visual analytical approach revealed spatially recognized differentially expressed genes that aligned with four gene clusters. Cluster 1 genes were present predominantly in DECs and Cluster 3 centered around CTC genes in all labor phenotypes. Cluster 2 genes were predominantly found in AECs in PPROM and PTB, while Cluster 4 contained AMC and CTC genes identified in term labor cases. We identified the top 10 differentially expressed genes and their connected pathways (kinase activation, NF-κB, inflammation, cytoskeletal remodeling, and hormone regulation) per cluster in each tissue layer. An in-depth understanding of the involvement of each system and cell layer may help provide targeted and tailored interventions to reduce the risk of PTB.


Asunto(s)
Decidua , Membranas Extraembrionarias , Nacimiento Prematuro , Transcriptoma , Femenino , Humanos , Embarazo , Decidua/metabolismo , Membranas Extraembrionarias/metabolismo , Nacimiento Prematuro/genética , Rotura Prematura de Membranas Fetales/genética , Rotura Prematura de Membranas Fetales/metabolismo , Nacimiento a Término/genética , Amnios/metabolismo , Amnios/citología , Adulto , Corion/metabolismo , Perfilación de la Expresión Génica
4.
Gynecol Endocrinol ; 40(1): 2382818, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39039858

RESUMEN

Premature ovarian insufficiency (POI) or premature ovarian failure (POF) is a multifactorial disorder occurring in reproductive-age women, characterized by elevated levels of follicle-stimulating hormone (FSH) and irregular or absent menstrual cycles, often accompanied by perimenopausal symptoms and infertility. While assisted reproductive technology can address the reproductive aspirations of some POI-affected women, it is hindered by issues such as exorbitant expenses, substantial risks, and poor rates of conception. Encouragingly, extensive research is exploring novel approaches to enhance fertility, particularly in the realm of stem cell therapy, showcasing both feasibility and significant potential. Human amniotic epithelial cells (hAECs) from discarded placental tissues are crucial in regenerative medicine for their pluripotency, low immunogenicity, non-tumorigenicity, accessibility, and minimal ethical concerns. Preclinical studies highlight the underlying mechanisms and therapeutic effects of hAECs in POI treatment, and current research is focusing on innovative interventions to augment hAECs' efficacy. However, despite these strides, overcoming application challenges is essential for successful clinical translation. This paper conducted a comprehensive analysis of the aforementioned issues, examining the prospects and challenges of hAECs in POI, with the aim of providing some insights for future research and clinical practice.


Asunto(s)
Amnios , Células Epiteliales , Insuficiencia Ovárica Primaria , Humanos , Insuficiencia Ovárica Primaria/terapia , Femenino , Células Epiteliales/trasplante , Amnios/citología , Amnios/trasplante
5.
Compend Contin Educ Dent ; 45(7): e1-e4, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39029967

RESUMEN

With increased awareness, both in the dental literature and by the general public, of peri-implant disease, a growing trend in dentistry is to save teeth with a "questionable" periodontal prognosis. This prospective study involving such patients was designed to evaluate the effects of combining a bioactive barrier and graft, not on the socket but to augment adjacent periodontal conditions on teeth with severe periodontal bone loss at the time of extraction of an adjacent tooth. Fifteen patients were selected; teeth were extracted, ground, prepared with a pH 11 cleanser, partially demineralized, and made into a graft. This mixture was used to augment socket volume and perform periodontal regenerative surgery. The graft was covered with a bioactive amnion-chorion barrier membrane. Bioactive membranes can stimulate host cells in the surrounding gingival and periosteal tissues to accelerate site closure and healing, simultaneously exerting positive effects on the underlying bone and graft material not observed to the same extent with other membranes. This can improve healing and site regeneration as shown clinically and radiographically in this report. Use of these bioactive barrier membrane and dentin graft materials may have additive effects and provide stimulus for conversion to host bone after site healing. The combination of an amnion-chorion membrane with autologous dentin graft appears to maximize the benefits of the individual materials, improving guided tissue regeneration results and the prognoses of periodontally involved teeth.


Asunto(s)
Amnios , Corion , Dentina , Extracción Dental , Humanos , Amnios/trasplante , Pronóstico , Estudios Prospectivos , Corion/trasplante , Persona de Mediana Edad , Femenino , Adulto , Masculino , Pérdida de Hueso Alveolar/cirugía , Regeneración Tisular Guiada Periodontal/métodos , Anciano
6.
Exp Clin Transplant ; 22(6): 451-454, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39072516

RESUMEN

OBJECTIVES: In this study, we aimed to define demographic data and trends in use of amniotic membrane transplant during the past decade at a tertiary eye center. MATERIALS AND METHODS: We included 272 patients who underwent amniotic membrane transplant for ocular surface pathology from January 2009 to December 2021. We retrospectively evaluated the medical data. RESULTS: The male-to-female ratio of patients was 41/23. Mean age of the patients was 50 ± 23.6 years (range, 1-91 years). Indications consisted of ocular surface lesion excision surgery (n = 184; 66.7%), chemical injury (n = 25; 9.1%), persistent epithelial defect (n = 23; 8.3%), keratitis (n = 22; 8%), noninfectious corneal perforation (n = 9; 3.3%), bullous keratopathy (n = 9; 3.3%), and ocular cicatricial pemphigoid (n = 4; 1.4%). Single amniotic membrane transplant was applied to 236 patients (85.5%), and multiple transplant was applied to 40 patients (14.5%). We observed repeated amniotic membrane transplant rates and amniotic membrane degradation durations that were associated with primary disease (P = .005 and P < .001, respectively). Degradation time was shorter in cases of chemical burns and keratitis than in cases after ocular surface lesion excision. Amniotic membrane transplant indication rates were statistically different between the first 6 years and the last 6 years of the 12 years of data (P = .041). The frequency of amniotic membrane transplant application in microbial keratitis has increased substantially in the past 2 years. CONCLUSIONS: Amniotic membrane is used as a biomaterial in various ocular surface diseases due to its anti-inflammatory, antimicrobial, and wound-healing properties. After transplant, the amniotic membrane, which is directly related to the inflam-matory processes of the primary disease, degrades gradually. There may be changes in the trend of amniotic membrane transplant, the indications of which are progressively expanding over time.


Asunto(s)
Amnios , Centros de Atención Terciaria , Humanos , Masculino , Femenino , Persona de Mediana Edad , Adulto , Amnios/trasplante , Estudios Retrospectivos , Anciano , Adolescente , Adulto Joven , Niño , Centros de Atención Terciaria/tendencias , Preescolar , Anciano de 80 o más Años , Resultado del Tratamiento , Lactante , Factores de Tiempo , Factores de Riesgo , Pautas de la Práctica en Medicina/tendencias , Oftalmopatías/cirugía , Turquía
7.
Sci Rep ; 14(1): 15998, 2024 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987293

RESUMEN

Pathological fibrosis is a significant complication of surgical procedures resulting from the accumulation of excess collagen at the site of repair which can compromise the tissue architecture and severely impede the function of the affected tissue. Few prophylactic treatments exist to counteract this process; however, the use of amniotic membrane allografts has demonstrated promising clinical outcomes. This study aimed to identify the underlying mechanism of action by utilizing relevant models that accurately represent the pathophysiology of the disease state. This study employed a pro-fibrotic in vitro system using TGFß1 stimulation and macromolecular crowding techniques to evaluate the mechanism by which amniotic membrane allografts regulate collagen biosynthesis and deposition. Following treatment with dehydrated human amnion chorion membrane (DHACM), subsequent RNA sequencing and functional enrichment with Reactome pathway analysis indicated that amniotic membranes are indeed capable of regulating genes associated with the composition and function of the extracellular matrix. Furthermore, macromolecular crowding was used in vitro to expand the evaluation to include both the effects of DHACM and a lyophilized human amnion/chorion membrane (LHACM). DHACM and LHACM regulate the TGFß pathway and myofibroblast differentiation. Additionally, both DHACM and LHACM modulate the production, secretion, and deposition of collagen type I, a primary target for pathological fibrosis. These observations support the hypothesis that amniotic membranes may interrupt pathological fibrosis by regulating collagen biosynthesis and associated pathways.


Asunto(s)
Amnios , Corion , Colágeno , Amnios/metabolismo , Humanos , Corion/metabolismo , Colágeno/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Diferenciación Celular , Matriz Extracelular/metabolismo , Miofibroblastos/metabolismo , Fibrosis , Femenino , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética
8.
J Wound Care ; 33(Sup7): S4-S14, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38973638

RESUMEN

OBJECTIVE: Diabetic foot ulcers (DFUs) continue to challenge wound care practitioners. This prospective, multicentre, randomised controlled trial (RCT) evaluated the effectiveness of a dehydrated Amnion Chorion Membrane (dACM) (Organogenesis Inc., US) versus standard of care (SoC) alone in complex DFUs in a challenging patient population. METHOD: Subjects with a DFU extending into dermis, subcutaneous tissue, tendon, capsule, bone or joint were enrolled in a 12-week trial. They were allocated equally to two treatment groups: dACM (plus SoC); or SoC alone. The primary endpoint was frequency of wound closure determined by a Cox analysis that adjusted for duration and wound area. Kaplan-Meier analysis was used to determine median time to complete wound closure (CWC). RESULTS: The cohort comprised 218 patients, and these were split equally between the two treatment groups with 109 patients in each. A Cox analysis showed that the estimated frequency of wound closure for the dACM plus SoC group was statistically superior to the SoC alone group at week 4 (12% versus 8%), week 6 (22% versus 11%), week 8 (31% versus 21%), week 10 (42% versus 27%) and week 12 (50% versus 35%), respectively (p=0.04). The computed hazard ratio (1.48 (confidence interval: 0.95, 2.29) showed a 48% greater probability of wound closure in favour of the dACM group. Median time to wound closure for dACM-treated ulcers was 84 days compared to 'not achieved' in the SoC-treated group (i.e., ≥50% of SoC-treated DFUs failed to heal by week 12; p=0.04). CONCLUSION: In an adequately powered DFU RCT, dACM increased the frequency, decreased the median time, and improved the probability of CWC when compared with SoC alone. dACM demonstrated beneficial effects in DFUs in a complex patient population. DECLARATION OF INTEREST: This study was funded by Organogenesis Inc., US. JC serves as a consultant and speaker for Organogenesis. RDD serves as a speaker for Organogenesis. OMA and MLS serve as consultants for Organogenesis. The authors have no other conflicts of interest to declare.


Asunto(s)
Amnios , Corion , Pie Diabético , Nivel de Atención , Cicatrización de Heridas , Humanos , Pie Diabético/terapia , Femenino , Amnios/trasplante , Masculino , Corion/trasplante , Persona de Mediana Edad , Estudios Prospectivos , Anciano , Resultado del Tratamiento , Adulto , Apósitos Biológicos
9.
Elife ; 122024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39051990

RESUMEN

Amniogenesis, a process critical for continuation of healthy pregnancy, is triggered in a collection of pluripotent epiblast cells as the human embryo implants. Previous studies have established that bone morphogenetic protein (BMP) signaling is a major driver of this lineage specifying process, but the downstream BMP-dependent transcriptional networks that lead to successful amniogenesis remain to be identified. This is, in part, due to the current lack of a robust and reproducible model system that enables mechanistic investigations exclusively into amniogenesis. Here, we developed an improved model of early amnion specification, using a human pluripotent stem cell-based platform in which the activation of BMP signaling is controlled and synchronous. Uniform amniogenesis is seen within 48 hr after BMP activation, and the resulting cells share transcriptomic characteristics with amnion cells of a gastrulating human embryo. Using detailed time-course transcriptomic analyses, we established a previously uncharacterized BMP-dependent amniotic transcriptional cascade, and identified markers that represent five distinct stages of amnion fate specification; the expression of selected markers was validated in early post-implantation macaque embryos. Moreover, a cohort of factors that could potentially control specific stages of amniogenesis was identified, including the transcription factor TFAP2A. Functionally, we determined that, once amniogenesis is triggered by the BMP pathway, TFAP2A controls the progression of amniogenesis. This work presents a temporally resolved transcriptomic resource for several previously uncharacterized amniogenesis states and demonstrates a critical intermediate role for TFAP2A during amnion fate specification.


Asunto(s)
Amnios , Proteínas Morfogenéticas Óseas , Regulación del Desarrollo de la Expresión Génica , Amnios/metabolismo , Amnios/embriología , Humanos , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Animales , Transducción de Señal , Perfilación de la Expresión Génica , Diferenciación Celular , Femenino , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Células Madre Pluripotentes/metabolismo , Embarazo
10.
Int J Mol Sci ; 25(13)2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-39000065

RESUMEN

Photochemical sealing of a nerve wrap over the repair site isolates and optimizes the regenerating nerve microenvironment. To facilitate clinical adoption of the technology, we investigated photosealed autologous tissue in a rodent sciatic nerve transection and repair model. Rats underwent transection of the sciatic nerve with repair performed in three groups: standard microsurgical neurorrhaphy (SN) and photochemical sealing with a crosslinked human amnion (xHAM) or autologous vein. Functional recovery was assessed at four-week intervals using footprint analysis. Gastrocnemius muscle mass preservation, histology, and nerve histomorphometry were evaluated at 120 days. Nerves treated with a PTB-sealed autologous vein improved functional recovery at 120 days although the comparison between groups was not significantly different (SN: -58.4 +/- 10.9; XHAM: -57.9 +/- 8.7; Vein: -52.4 +/- 17.1). Good muscle mass preservation was observed in all groups, with no statistical differences between groups (SN: 69 +/- 7%; XHAM: 70 +/- 7%; Vein: 70 +/- 7%). Histomorphometry showed good axonal regeneration in all repair techniques. These results demonstrate that peripheral nerve repair using photosealed autologous veins produced regeneration at least equivalent to current gold-standard microsurgery. The use of autologous veins removes costs and foreign body concerns and would be readily available during surgery. This study illustrates a new repair method that could restore normal endoneurial homeostasis with minimal trauma following severe nerve injury.


Asunto(s)
Regeneración Nerviosa , Nervio Ciático , Animales , Ratas , Regeneración Nerviosa/fisiología , Nervio Ciático/lesiones , Nervio Ciático/cirugía , Nervio Ciático/fisiología , Humanos , Amnios , Trasplante Autólogo/métodos , Músculo Esquelético , Recuperación de la Función , Masculino , Procedimientos Neuroquirúrgicos/métodos , Venas/cirugía
11.
Biomed Microdevices ; 26(3): 32, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38963644

RESUMEN

Fetal membrane (amniochorion), the innermost lining of the intrauterine cavity, surround the fetus and enclose amniotic fluid. Unlike unidirectional blood flow, amniotic fluid subtly rocks back and forth, and thus, the innermost amnion epithelial cells are continuously exposed to low levels of shear stress from fluid undulation. Here, we tested the impact of fluid motion on amnion epithelial cells (AECs) as a bearer of force impact and their potential vulnerability to cytopathologic changes that can destabilize fetal membrane functions. A previously developed amnion membrane (AM) organ-on-chip (OOC) was utilized but with dynamic flow to culture human fetal amnion membrane cells. The applied flow was modulated to perfuse culture media back and forth for 48 h to mimic fluid motion. A static culture condition was used as a negative control, and oxidative stress (OS) condition was used as a positive control representing pathophysiological changes. The impacts of fluidic motion were evaluated by measuring cell viability, cellular transition, and inflammation. Additionally, scanning electron microscopy (SEM) imaging was performed to observe microvilli formation. The results show that regardless of the applied flow rate, AECs and AMCs maintained their viability, morphology, innate meta-state, and low production of pro-inflammatory cytokines. E-cadherin expression and microvilli formation in the AECs were upregulated in a flow rate-dependent fashion; however, this did not impact cellular morphology or cellular transition or inflammation. OS treatment induced a mesenchymal morphology, significantly higher vimentin to cytokeratin 18 (CK-18) ratio, and pro-inflammatory cytokine production in AECs, whereas AMCs did not respond in any significant manner. Fluid motion and shear stress, if any, did not impact AEC cell function and did not cause inflammation. Thus, when using an amnion membrane OOC model, the inclusion of a dynamic flow environment is not necessary to mimic in utero physiologic cellular conditions of an amnion membrane.


Asunto(s)
Líquido Amniótico , Membranas Extraembrionarias , Dispositivos Laboratorio en un Chip , Humanos , Líquido Amniótico/citología , Membranas Extraembrionarias/citología , Membranas Extraembrionarias/metabolismo , Amnios/citología , Amnios/metabolismo , Supervivencia Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Movimiento (Física) , Estrés Oxidativo , Modelos Biológicos , Sistemas Microfisiológicos
12.
Sci Rep ; 14(1): 17407, 2024 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-39075142

RESUMEN

Currently, in vitro cultured corneal epithelial transplantation is effective in treating limbal stem cell dysfunction (LSCD). Selecting carriers is crucial for constructing the corneal epithelium through tissue engineering. In this study, the traditional amniotic membrane (AM) was modified, and mesenchymal stem cells (MSCs) were inoculated into the ultra-thin amniotic membrane (UAM) stroma to construct a novel UAM-MSC tissue-engineered corneal epithelial carrier, that could effectively simulate the limbal stem cells (LSCs) microenvironment. The structure of different carriers cultured tissue-engineered corneal epithelium and the managed rabbit LSCD model corneas were observed through hematoxylin-eosin staining. Cell phenotypes were evaluated through fluorescence staining, Western blotting, and RT-qPCR. Additionally, cell junction genes and expression markers related to anti-neovascularization were evaluated using RT-qPCR. Corneal epithelium cell junctions were observed via an electron microscope. The tissue-engineered corneal epithelium culture medium was analyzed through mass spectrometry. Tissue-engineered corneal epithelial cells expanded by LSCs on UAM-MSCs had good transparency. Simultaneously, progenitor cell (K14, PNCA, p63) and corneal epithelial (PAX6) gene expression in tissue-engineered corneal epithelium constructed using UAM-MSCs was higher than that in corneal epithelial cells amplified by UAM and de-epithelialized amniotic membrane. Electron microscopy revealed that corneal epithelial cells grafted with UAM-MSCs were closely connected. In conclusion, the UAM-MSCs vector we constructed could better simulate the limbal microenvironment; the cultured tissue-engineered corneal epithelium had better transparency, anti-neovascularization properties, closer intercellular connections, and closer resemblance to the natural corneal epithelial tissue phenotype.


Asunto(s)
Amnios , Epitelio Corneal , Células Madre Mesenquimatosas , Ingeniería de Tejidos , Amnios/citología , Ingeniería de Tejidos/métodos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Epitelio Corneal/citología , Epitelio Corneal/metabolismo , Animales , Conejos , Humanos , Células Cultivadas , Limbo de la Córnea/citología , Limbo de la Córnea/metabolismo , Diferenciación Celular
13.
Acta Obstet Gynecol Scand ; 103(9): 1829-1837, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38973223

RESUMEN

INTRODUCTION: Treatment of oligohydramnios in the mid-trimester is challenging, because of the high incidence of adverse perinatal outcomes mainly due to bronchopulmonary dysplasia. Antenatal amnioinfusion has been proposed as a possible treatment for oligohydramnios with intact amnions, but there are few relevant studies. This study aimed to evaluate the effectiveness of transabdominal amnioinfusion in the management of oligohydramnios without fetal lethal malformations in the second and early third trimesters. MATERIAL AND METHODS: It is a historical cohort study. A total of 79 patients diagnosed with oligohydramnios at 18-32 weeks gestation were enrolled. In the amnioinfusion group (n = 39), patients received transabdominal amnioinfusion with the assistance of real-time ultrasound guidance. In the expectant group (n = 41), patients were treated with 3000 mL of intravenous isotonic fluids daily. The perioperative complications and perinatal outcomes were analyzed. RESULTS: Compared with the expectant group, the delivery latency was significantly prolonged, and the rate of cesarean delivery was significantly reduced in the amnioinfusion group (p < 0.05). Although the rate of intrauterine fetal death was significantly reduced, the incidence of spontaneous miscarriage, premature rupture of membranes (PROMs), and threatened preterm labor were significantly higher in the amnioinfusion group than in the expectant group (p < 0.05). There was no significant difference in terms of perinatal mortality (28.9% vs. 41.4%, p > 0.05). Multivariate logistic regression revealed that amnioinfusion (odds ratio [OR] 0.162, 95% confidence interval [CI] 0.04-0.61, p = 0.008) and gestational age at diagnosis (OR 0.185, 95% CI 0.04-0.73, p = 0.016) were independently associated with neonatal adverse outcomes. Further subgrouping showed that amnioinfusion significantly reduced the frequency of bronchopulmonary hypoplasia for patients ≤26 weeks (26.7% vs. 75.0%, p = 0.021). The rates of other neonatal complications were similar in both groups. CONCLUSIONS: Amnioinfusion has no significant effect on improving the perinatal mortality of oligohydramnios in the second and early third trimesters. It may lead to a relatively high rate of PROM and spontaneous abortion. However, amnioinfusion may significantly improve the latency period, the rate of cesarean delivery, and neonatal outcomes of oligohydramnios, especially for women ≤26 weeks with high risk of neonatal bronchopulmonary hypoplasia.


Asunto(s)
Oligohidramnios , Segundo Trimestre del Embarazo , Tercer Trimestre del Embarazo , Humanos , Femenino , Oligohidramnios/terapia , Embarazo , Adulto , Líquido Amniótico , Resultado del Embarazo , Recién Nacido , Estudios de Cohortes , Espera Vigilante , Cesárea , Resultado del Tratamiento , Edad Gestacional , Amnios , Ultrasonografía Prenatal
14.
Biomater Sci ; 12(17): 4393-4406, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39034884

RESUMEN

The ordered arrangement of cells and extracellular matrix facilitates the seamless transmission of electrical signals along axons in the spinal cord and peripheral nerves. Therefore, restoring tissue geometry is crucial for neural regeneration. This study presents a novel method using proteins derived from the human amniotic membrane, which is modified with photoresponsive groups, to produce cryogels with aligned porosity. Freeze-casting was used to produce cryogels with longitudinally aligned pores, while cryogels with randomly distributed porosity were used as the control. The cryogels exhibited remarkable injectability and shape-recovery properties, essential for minimally invasive applications. Different tendencies in proliferation and differentiation were evident between aligned and random cryogels, underscoring the significance of the scaffold's microstructure in directing the behaviour of neural stem cells (NSC). Remarkably, aligned cryogels facilitated extensive cellular infiltration and migration, contrasting with NSC cultured on isotropic cryogels, which predominantly remained on the scaffold's surface throughout the proliferation experiment. Significantly, the proliferation assay demonstrated that on day 7, the aligned cryogels contained eight times more cells compared to the random cryogels. Consistent with the proliferation experiments, NSC exhibited the ability to differentiate into neurons within the aligned scaffolds and extend neurites longitudinally. In addition, differentiation assays showed a four-fold increase in the expression of neural markers in the cross-sections of the aligned cryogels. Conversely, the random cryogels exhibited minimal presence of cell bodies and extensions. The presence of synaptic vesicles on the anisotropic cryogels indicates the formation of functional synaptic connections, emphasizing the importance of the scaffold's microstructure in guiding neuronal reconnection.


Asunto(s)
Amnios , Diferenciación Celular , Proliferación Celular , Criogeles , Regeneración Nerviosa , Células-Madre Neurales , Andamios del Tejido , Amnios/química , Criogeles/química , Humanos , Células-Madre Neurales/citología , Andamios del Tejido/química , Animales , Porosidad , Ingeniería de Tejidos , Células Cultivadas
15.
Endocrinology ; 165(9)2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39082703

RESUMEN

Premature rupture of membranes (PROM) is defined as rupture of fetal membranes before the onset of labor. Prolactin (PRL) is secreted by decidual membranes and accumulated significantly in the amniotic fluid during pregnancy. PRL could ameliorate inflammation and collagen degradation in fetal membranes. However, the role of PRL in amniotic membrane is not well characterized. We isolated human amniotic epithelial stem cells (hAESCs) from human fetal membranes to study the effect of PRL on proliferation, migration, and antioxidative stress. Amniotic pore culture technique (APCT) model was constructed to evaluate the tissue regeneration effect in vitro. The potential targets and pathways of PRL acting in amnion via integrated bioinformatic methods. PRL had a dose-dependent effect on hAESCs in vitro. PRL (500 ng/mL) significantly improved the viability of hAESCs and inhibited cell apoptosis, related to the upregulation of CCN2 expression and downregulation of Bax, Caspase 3, and Caspase 8. PRL accelerated migration process in hAESCs via downregulation of MMP2, MMP3, and MMP9. PRL attenuated the cellular damage and mitochondrial dysfunction induced by hydrogen peroxide in hAESCs. PRL accelerated the healing process in the APCT model significantly. The top 10 specific targets (IGF1R, SIRT1, MAP2K1, CASP8, MAPK14, MCL1, NFKB1, HIF1A, MTOR, and HSP90AA1) and signaling pathways (such as HIF signaling pathway) were selected using an integrated bioinformatics approach. PRL improves the viability and antioxidative stress function of hAESCs and the regeneration of ruptured amniotic membranes in vitro. Thus, PRL has great therapeutic potential for prevention and treatment of ruptured membranes.


Asunto(s)
Amnios , Apoptosis , Rotura Prematura de Membranas Fetales , Prolactina , Humanos , Amnios/metabolismo , Amnios/citología , Rotura Prematura de Membranas Fetales/terapia , Rotura Prematura de Membranas Fetales/metabolismo , Prolactina/metabolismo , Prolactina/farmacología , Femenino , Embarazo , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Regeneración/fisiología , Regeneración/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Células Epiteliales/efectos de los fármacos , Células Madre/metabolismo , Supervivencia Celular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
16.
PeerJ ; 12: e17616, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38952966

RESUMEN

Background: Mesenchymal stem cells (MSCs) are increasingly recognized for their regenerative potential. However, their clinical application is hindered by their inherent variability, which is influenced by various factors, such as the tissue source, culture conditions, and passage number. Methods: MSCs were sourced from clinically relevant tissues, including adipose tissue-derived MSCs (ADMSCs, n = 2), chorionic villi-derived MSCs (CMMSCs, n = 2), amniotic membrane-derived MSCs (AMMSCs, n = 3), and umbilical cord-derived MSCs (UCMSCs, n = 3). Passages included the umbilical cord at P0 (UCMSCP0, n = 2), P3 (UCMSCP3, n = 2), and P5 (UCMSCP5, n = 2) as well as the umbilical cord at P5 cultured under low-oxygen conditions (UCMSCP5L, n = 2). Results: We observed that MSCs from different tissue origins clustered into six distinct functional subpopulations, each with varying proportions. Notably, ADMSCs exhibited a higher proportion of subpopulations associated with vascular regeneration, suggesting that they are beneficial for applications in vascular regeneration. Additionally, CMMSCs had a high proportion of subpopulations associated with reproductive processes. UCMSCP5 and UCMSCP5L had higher proportions of subpopulations related to female reproductive function than those for earlier passages. Furthermore, UCMSCP5L, cultured under low-oxygen (hypoxic) conditions, had a high proportion of subpopulations associated with pro-angiogenic characteristics, with implications for optimizing vascular regeneration. Conclusions: This study revealed variation in the distribution of MSC subpopulations among different tissue sources, passages, and culture conditions, including differences in functions related to vascular and reproductive system regeneration. These findings hold promise for personalized regenerative medicine and may lead to more effective clinical treatments across a spectrum of medical conditions.


Asunto(s)
Tejido Adiposo , Células Madre Mesenquimatosas , Cordón Umbilical , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Humanos , Cordón Umbilical/citología , Femenino , Tejido Adiposo/citología , Células Cultivadas , Vellosidades Coriónicas/fisiología , Amnios/citología , Diferenciación Celular
17.
Int J Mol Sci ; 25(13)2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-39000523

RESUMEN

The dental implant surface plays a crucial role in osseointegration. The topography and physicochemical properties will affect the cellular functions. In this research, four distinct titanium surfaces have been studied: machined acting (MACH), acid etched (AE), grit blasting (GBLAST), and a combination of grit blasting and subsequent acid etching (GBLAST + AE). Human amniotic mesenchymal (hAMSCs) and epithelial stem cells (hAECs) isolated from the amniotic membrane have attractive stem-cell properties. They were cultured on titanium surfaces to analyze their impact on biological behavior. The surface roughness, microhardness, wettability, and surface energy were analyzed using interferometric microscopy, Vickers indentation, and drop-sessile techniques. The GBLAST and GBLAST + AE surfaces showed higher roughness, reduced hydrophilicity, and lower surface energy with significant differences. Increased microhardness values for GBLAST and GBLAST + AE implants were attributed to surface compression. Cell viability was higher for hAMSCs, particularly on GBLAST and GBLAST + AE surfaces. Alkaline phosphatase activity enhanced in hAMSCs cultured on GBLAST and GBLAST + AE surfaces, while hAECs showed no mineralization signals. Osteogenic gene expression was upregulated in hAMSCs on GBLAST surfaces. Moreover, α2 and ß1 integrin expression enhanced in hAMSCs, suggesting a surface-integrin interaction. Consequently, hAMSCs would tend toward osteoblastic differentiation on grit-blasted surfaces conducive to osseointegration, a phenomenon not observed in hAECs.


Asunto(s)
Amnios , Implantes Dentales , Propiedades de Superficie , Titanio , Humanos , Titanio/química , Amnios/citología , Amnios/metabolismo , Osteogénesis , Diferenciación Celular , Células Cultivadas , Oseointegración , Células Madre/citología , Células Madre/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Supervivencia Celular , Fosfatasa Alcalina/metabolismo
18.
Skin Res Technol ; 30(7): e13860, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39073182

RESUMEN

BACKGROUND: The perfect repair of damaged skin has always been a constant goal for scientists; however, the repair and reconstruction of skin is still a major problem and challenge in injury and burns medicine. Human amniotic membrane (hAM), with its good mechanical properties and anti-inflammatory, antioxidant and antimicrobial benefits, containing growth factors that promote wound healing, has evolved over the last few decades from simple skin sheets to high-tech dressings, such as being made into nanocomposites, hydrogels, powders, and electrostatically spun scaffolds. This paper aims to explore the historical development, applications, trends, and research hotspots of hAM in wound healing. METHODS: We examined 2660 publications indexed in the Web of Science Core Collection (WoSCC) from January 1, 1975 to July 12, 2023. Utilizing bibliometric methods, we employed VOSviewer, CiteSpace, and R-bibliometrix to characterize general information, identify development trends, and highlight research hotspots. Subsequently, we identified a collection of high-quality English articles focusing on the roles of human amniotic epithelial stem cells (hAESCs), human amniotic mesenchymal stem cells (hAMSCs), and amniotic membrane (AM) scaffolds in regenerative medicine and tissue engineering. RESULTS: Bibliometric analysis identified Udice-French Research Universities as the most productive affiliation and Tseng S.C.G. as the most prolific author. Keyword analysis, historical direct quotations network, and thematic analysis helped us review the historical and major themes in this field. Our examination included the knowledge structure, global status, trends, and research hotspots regarding the application of hAM in wound healing. Our findings indicate that contemporary research emphasizes the preparation and application of products derived from hAM. Notably, both hAM and the cells isolated from it - hADSCs and hAESCs are prominent and promising areas of research in regenerative medicine and tissue engineering. CONCLUSION: This research delivers a comprehensive understanding of the knowledge frameworks, global dynamics, emerging patterns, and primary research foci in the realm of hAM applications for wound healing. The field is rapidly evolving, and our findings offer valuable insights for researchers. Future research outcomes are anticipated to be applied in clinical practice, enhancing methods for disease prevention, diagnosis, and treatment.


Asunto(s)
Amnios , Cicatrización de Heridas , Humanos , Ingeniería de Tejidos/métodos , Apósitos Biológicos , Andamios del Tejido , Células Epiteliales/fisiología
19.
Stem Cell Rev Rep ; 20(6): 1618-1635, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38831179

RESUMEN

Autoimmune factors play an important role in premature ovarian insufficiency (POI). Human amniotic epithelial stem cells (hAESCs) have recently shown promising treatment effects on chemotherapy-induced POI. However, the therapeutic efficacy and underlying mechanisms of hAESCs in autoimmune POI remain to be investigated. In this study, we showed for the first time that intravenous transplantation of hAESCs could reside in the ovary of zona pellucida 3 peptide (pZP3) induced autoimmune POI mice model for at least 4 weeks. hAESCs could improve ovarian function and fertility, alleviate inflammation and reduce apoptosis of granulosa cells (GCs) in autoimmune POI mice. The transcriptome analysis of mice ovaries and in vitro co-cultivation experiments suggest that activation of the AKT and ERK pathways may be the key mechanism in the therapeutic effect of hAESCs. Our work provides the theoretical and experimental foundation for optimizing the administration of hAESCs, as well as the clinical application of hAESCs in autoimmune POI patients.


Asunto(s)
Amnios , Células Epiteliales , Células de la Granulosa , Sistema de Señalización de MAP Quinasas , Insuficiencia Ovárica Primaria , Proteínas Proto-Oncogénicas c-akt , Células Madre , Femenino , Animales , Insuficiencia Ovárica Primaria/terapia , Insuficiencia Ovárica Primaria/patología , Insuficiencia Ovárica Primaria/metabolismo , Humanos , Ratones , Amnios/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células de la Granulosa/metabolismo , Células Epiteliales/metabolismo , Células Madre/metabolismo , Células Madre/citología , Modelos Animales de Enfermedad , Trasplante de Células Madre , Apoptosis , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/patología , Glicoproteínas de la Zona Pelúcida/metabolismo , Glicoproteínas de la Zona Pelúcida/genética
20.
Stem Cells Transl Med ; 13(8): 711-723, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-38895873

RESUMEN

Lung and brain injury that occurs during the perinatal period leads to lifelong disability and is often driven and/or exacerbated by inflammation. Human amniotic epithelial cells (hAEC), which demonstrate immunomodulatory, anti-fibrotic, and regenerative capabilities, are being explored as a therapeutic candidate for perinatal injury. However, limitations regarding scalable manufacturing, storage, transport, and dose-related toxicity have impeded clinical translation. Isolated therapeutic extracellular vesicles (EVs) from stem and stem-like cells are thought to be key paracrine mediators of therapeutic efficacy. The unique characteristics of EVs suggest that they potentially circumvent the limitations of traditional cell-based therapies. However, given the novelty of EVs as a therapeutic, recommendations around ideal methods of production, isolation, storage, and delivery have not yet been created by regulatory agencies. In this concise review, we discuss the pertinence and limitations of cell-based therapeutics in perinatal medicine. We also review the preclinical evidence supporting the use of therapeutic EVs for perinatal therapy. Further, we summarize the arising considerations regarding adequate cell source, biodistribution, isolation and storage methods, and regulatory roadblocks for the development of therapeutic EVs.


Asunto(s)
Amnios , Células Epiteliales , Vesículas Extracelulares , Humanos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Células Epiteliales/citología , Células Epiteliales/metabolismo , Amnios/citología , Lesiones Encefálicas/terapia , Lesión Pulmonar/terapia , Animales , Femenino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA